1
|
Xiao Q, Li Y, Cai B, Huang X, Fang L, Liang F, Chen L, Xu K, Zhang W, Wang X, Yin A, Wang X, Cai Z, Zhuang F, Shao Q, Zhou B, Hocher B, He B, Shen L. CCDC80 Protects against Aortic Dissection and Rupture by Maintaining the Contractile Smooth Muscle Cell Phenotype. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2502108. [PMID: 40278823 DOI: 10.1002/advs.202502108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/20/2025] [Indexed: 04/26/2025]
Abstract
Aortic dissection (AD) is a life-threatening medical emergency characterized by adverse vascular remodeling. Coiled-coil domain-containing protein 80 (CCDC80) plays an essential role in regulating cardiovascular remodeling. This study aims to define the role of CCDC80 in the formation and development of AD. Significant downregulation of CCDC80 in vascular smooth muscle cell (VSMC) in human and mouse AD is identified. Then, CCDC80 knockout mice (CCDC80-/-) and VSMC-specific CCDC80 knockout mice (CCDC80fl/fl SM22α Cre+) treated with angiotensin II (Ang II) or Ang II combined with β-aminopropionitrile monofumarate (BAPN) frequently develop AD with higher frequency and severity, accompanied by severe elastin fragmentation and collagen deposition. Mechanistically, CCDC80 interacts with JAK2, and CCDC80 deficiency promotes VSMC phenotype switching, proliferation, and migration as well as matrix metalloproteinase production by activating the JAK2/STAT3 signaling pathway. Moreover, the JAK2/STAT3 pathway-specific inhibitor ameliorates adverse vascular remodeling and reduces AD formation in CCDC80-knockout mice by mitigating VSMC phenotype switching. In conclusion, CCDC80 deficiency exacerbates the progression of events leading to AD by activating the JAK2/STAT3 pathway involved in regulating the phenotype switching and function of VSMCs. These findings highlight that CCDC80 is a potential key target for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Qingqing Xiao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Bin Cai
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiying Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Liang Fang
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Long Chen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ke Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Weifeng Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xiaolei Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Anwen Yin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Fei Zhuang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Qin Shao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Bin Zhou
- Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200032, China
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, 69123, Heidelberg, Germany
- Reproductive and Genetic Hospital of CITIC-Xiangya, People's Republic of China, Changsha, 410028, China
- IMD Institut fur Medizinische Diagnostik Berlin-Potsdam GbR, 14473, Berlin, Germany
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| |
Collapse
|
2
|
Wang H, Kazaleh M, Gioscia-Ryan R, Millar J, Temprano-Sagrera G, Wood S, Van Den Bergh F, Blin MG, Wragg KM, Luna A, Hawkins RB, Soleimanpour SA, Sabater-Lleal M, Shu C, Beard DA, Ailawadi G, Deng JC, Goldstein DR, Salmon M. Deficiency of mitophagy mediator Parkin in aortic smooth muscle cells exacerbates abdominal aortic aneurysm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.30.621201. [PMID: 39554010 PMCID: PMC11565987 DOI: 10.1101/2024.10.30.621201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Abdominal aortic aneurysms (AAAs) are a degenerative aortic disease and associated with hallmarks of aging, such as mitophagy. Despite this, the exact associations among mitophagy, aging, and AAA progression remain unknown. In our study, gene expression analysis of human AAA tissue revealed downregulation of mitophagy pathways, mitochondrial structure, and function-related proteins. Human proteomic analyses identified decreased levels of mitophagy mediators PINK1 and Parkin. Aged mice and, separately, a murine AAA model showed reduced mitophagy in aortic vascular smooth muscle cells (VSMCs) and PINK1 and Parkin expression. Parkin knockdown in VSMCs aggravated AAA dilation in murine models, with elevated mitochondrial ROS and impaired mitochondrial function. Importantly, inhibiting USP30, an antagonist of the PINK1/Parkin pathway, increased mitophagy in VSMCs, improved mitochondrial function, and reduced AAA incidence and growth. Our study elucidates a critical mechanism that proposes AAAs as an age-associated disease with altered mitophagy, introducing new potential therapeutic approaches.
Collapse
|
3
|
Laloo R, Bailey M. TRPM7 channel as a potential therapeutic target for AAA. NATURE CARDIOVASCULAR RESEARCH 2025; 4:126-128. [PMID: 39953274 DOI: 10.1038/s44161-024-00598-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Affiliation(s)
- Ryan Laloo
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK.
- Leeds Vascular Institute, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| | - Marc Bailey
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
- Leeds Vascular Institute, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| |
Collapse
|
4
|
Zong P, Li CX, Feng J, Yue Z, Nethramangalath T, Xie Y, Qin X, Cicchetti M, Cai Y, Jellison E, Matsushita M, Runnels LW, Yue L. TRPM7 channel activity promotes the pathogenesis of abdominal aortic aneurysms. NATURE CARDIOVASCULAR RESEARCH 2025; 4:197-215. [PMID: 39953276 DOI: 10.1038/s44161-024-00596-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 12/04/2024] [Indexed: 02/17/2025]
Abstract
Abdominal aortic aneurysms (AAAs) occur in 1-2% of the elderly. The rupture of an AAA usually causes uncontrollable lethal hemorrhage, and its risk increases with AAA size. However, there is no effective pharmacological therapy for hindering AAA growth. Here we show that global or vascular smooth muscle cell (VSMC)-specific transient receptor potential melastatin 7 (TRPM7) knockout in mice prevented AAA formation, as indicated by inhibited VSMC reprogramming, reduced inflammatory infiltration and suppressed matrix degradation. Mechanistically, we showed that TRPM7-mediated Ca2+ signaling promotes Kruppel-like factor 4 (KLF4) activation, driving VSMC reprogramming and accelerating AAA growth. By generating channel-dead and using kinase-inactive knockin mice, we found that it is the channel function, rather than kinase activity, that is required for TRPM7-mediated AAA pathogenesis. Importantly, TRPM7 inhibitor NS8593 suppressed VSMC reprogramming and protected mice against AAA formation. Our data suggest that TRPM7 is a promising therapeutic target for developing effective prophylactic medications to limit AAA progression. In addition, the channel-dead TRPM7 knockin mice will serve as a valuable tool for elucidating the roles of TRPM7 in other pathophysiological conditions.
Collapse
MESH Headings
- Animals
- TRPM Cation Channels/genetics
- TRPM Cation Channels/metabolism
- TRPM Cation Channels/deficiency
- TRPM Cation Channels/antagonists & inhibitors
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/prevention & control
- Mice, Knockout
- Disease Models, Animal
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Kruppel-Like Factor 4
- Male
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/deficiency
- Mice, Inbred C57BL
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Calcium Signaling/drug effects
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/drug effects
- Humans
- Cells, Cultured
- Vascular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA
| | - Cindy X Li
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Zhichao Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | | | - Yangzhouyun Xie
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Xin Qin
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Mara Cicchetti
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Yujun Cai
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Evan Jellison
- Department of Immunology, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Masayuki Matsushita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Loren W Runnels
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA.
| |
Collapse
|
5
|
Wu H, Li Z, Yang L, He L, Liu H, Yang S, Xu Q, Li Y, Li W, Li Y, Gong Z, Shen Y, Yang X, Huang J, Yu F, Li L, Zhu J, Sun L, Fu Y, Kong W. ANK Deficiency-Mediated Cytosolic Citrate Accumulation Promotes Aortic Aneurysm. Circ Res 2024; 135:1175-1192. [PMID: 39513269 DOI: 10.1161/circresaha.124.325152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Disturbed metabolism and transport of citrate play significant roles in various pathologies. However, vascular citrate regulation and its potential role in aortic aneurysm (AA) development remain poorly understood. METHODS Untargeted metabolomics by mass spectrometry was applied to identify upregulated metabolites of the tricarboxylic acid cycle in AA tissues of mice. To investigate the role of citrate and its transporter ANK (progressive ankylosis protein) in AA development, vascular smooth muscle cell (VSMC)-specific Ank-knockout mice were used in both Ang II (angiotensin II)- and CaPO4-induced AA models. RESULTS Citrate was abnormally increased in both human and murine aneurysmal tissues, which was associated with downregulation of ANK, a citrate membrane transporter, in VSMCs. The knockout of Ank in VSMCs promoted AA formation in both Ang II- and CaPO4-induced AA models, while its overexpression inhibited the development of aneurysms. Mechanistically, ANK deficiency in VSMCs caused abnormal cytosolic accumulation of citrate, which was cleaved into acetyl coenzyme A and thus intensified histone acetylation at H3K23, H3K27, and H4K5. Cleavage under target and tagmentation analysis further identified that ANK deficiency-induced histone acetylation activated the transcription of inflammatory genes in VSMCs and thus promoted a citrate-related proinflammatory VSMC phenotype during aneurysm diseases. Accordingly, suppressing citrate cleavage to acetyl coenzyme A downregulated inflammatory gene expression in VSMCs and restricted ANK deficiency-aggravated AA formation. CONCLUSIONS Our studies define the pathogenic role of ANK deficiency-induced cytosolic citrate accumulation in AA pathogenesis and an undescribed citrate-related proinflammatory VSMC phenotype. Targeting ANK-mediated citrate transport may emerge as a novel diagnostic and therapeutic strategy in AA.
Collapse
MESH Headings
- Animals
- Mice
- Citric Acid/metabolism
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Aortic Aneurysm/etiology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Inbred C57BL
- Cytosol/metabolism
- Male
- Cells, Cultured
- Acetylation
- Acetyl Coenzyme A/metabolism
- Disease Models, Animal
- Histones/metabolism
Collapse
Affiliation(s)
- Hao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China (L.Y.)
| | - Lin He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hao Liu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, China (H.L., Q.X., J.Z.)
| | - Shiyu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qinfeng Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yanjie Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenqiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yiran Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
- Hwamei College of Life and Health Sciences, Zhejiang Wanli University, Ningbo, China (Z.G.)
| | - Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xueyuan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Li Li
- Department of Pathology, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.L.)
| | - Junming Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Luyang Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | | |
Collapse
|
6
|
Wang B, Cui K, Zhu B, Dong Y, Wang D, Singh B, Wu H, Li K, Eisa-Beygi S, Sun Y, Wong S, Cowan DB, Chen Y, Du M, Chen H. Epsins oversee smooth muscle cell reprograming by influencing master regulators KLF4 and OCT4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574714. [PMID: 39131381 PMCID: PMC11312448 DOI: 10.1101/2024.01.08.574714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Smooth muscle cells in major arteries play a crucial role in regulating coronary artery disease. Conversion of smooth muscle cells into other adverse cell types in the artery propels the pathogenesis of the disease. Curtailing artery plaque buildup by modulating smooth muscle cell reprograming presents us a new opportunity to thwart coronary artery disease. Here, our report how Epsins, a family of endocytic adaptor proteins oversee the smooth muscle cell reprograming by influencing master regulators OCT4 and KLF4. Using single-cell RNA sequencing, we characterized the phenotype of modulated smooth muscle cells in mouse atherosclerotic plaque and found that smooth muscle cells lacking epsins undergo profound reprogramming into not only beneficial myofibroblasts but also endothelial cells for injury repair of diseased endothelium. Our work lays concrete groundwork to explore an uncharted territory as we show that depleting Epsins bolsters smooth muscle cells reprograming to endothelial cells by augmenting OCT4 activity but restrain them from reprograming to harmful foam cells by destabilizing KLF4, a master regulator of adverse reprograming of smooth muscle cells. Moreover, the expression of Epsins in smooth muscle cells positively correlates with the severity of both human and mouse coronary artery disease. Integrating our scRNA-seq data with human Genome-Wide Association Studies (GWAS) identifies pivotal roles Epsins play in smooth muscle cells in the pathological process leading to coronary artery disease. Our findings reveal a previously unexplored direction for smooth muscle cell phenotypic modulation in the development and progression of coronary artery disease and unveil Epsins and their downstream new targets as promising novel therapeutic targets for mitigating metabolic disorders.
Collapse
Affiliation(s)
- Beibei Wang
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Kui Cui
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Bo Zhu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yunzhou Dong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Donghai Wang
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Bandana Singh
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Hao Wu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Kathryn Li
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yong Sun
- Department of Pathology, Birmingham, AL 35294, USA; University of Alabama at Birmingham, and the Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA
| | - Scott Wong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Douglas B. Cowan
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yabing Chen
- Department of Pathology, Birmingham, AL 35294, USA; University of Alabama at Birmingham, and the Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA
| | - Mulong Du
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
7
|
Quelquejay H, Al-Rifai R, Silvestro M, Vandestienne M, Ferreira I, Mirault T, Henrion D, Zhong X, Santos-Zas I, Goudot G, Alayrac P, Robidel E, Autret G, Balvay D, Taleb S, Tedgui A, Boulanger CM, Zernecke A, Saliba AE, Hadchouel J, Ramkhelawon B, Cochain C, Bergaya S, Jeunemaitre X, Ait-Oufella H. L-Wnk1 Deletion in Smooth Muscle Cells Causes Aortitis and Inflammatory Shift. Circ Res 2024; 135:488-502. [PMID: 38979610 DOI: 10.1161/circresaha.124.324366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND The long isoform of the Wnk1 (with-no-lysine [K] kinase 1) is a ubiquitous serine/threonine kinase, but its role in vascular smooth muscle cells (VSMCs) pathophysiology remains unknown. METHODS AngII (angiotensin II) was infused in Apoe-/- to induce experimental aortic aneurysm. Mice carrying an Sm22-Cre allele were cross-bred with mice carrying a floxed Wnk1 allele to specifically investigate the functional role of Wnk1 in VSMCs. RESULTS Single-cell RNA-sequencing of the aneurysmal abdominal aorta from AngII-infused Apoe-/- mice revealed that VSMCs that did not express Wnk1 showed lower expression of contractile phenotype markers and increased inflammatory activity. Interestingly, WNK1 gene expression in VSMCs was decreased in human abdominal aortic aneurysm. Wnk1-deficient VSMCs lost their contractile function and exhibited a proinflammatory phenotype, characterized by the production of matrix metalloproteases, as well as cytokines and chemokines, which contributed to local accumulation of inflammatory macrophages, Ly6Chi monocytes, and γδ T cells. Sm22Cre+Wnk1lox/lox mice spontaneously developed aortitis in the infrarenal abdominal aorta, which extended to the thoracic area over time without any negative effect on long-term survival. AngII infusion in Sm22Cre+Wnk1lox/lox mice aggravated the aortic disease, with the formation of lethal abdominal aortic aneurysms. Pharmacological blockade of γδ T-cell recruitment using neutralizing anti-CXCL9 (anti-CXC motif chemokine ligand 9) antibody treatment, or of monocyte/macrophage using Ki20227, a selective inhibitor of CSF1 receptor, attenuated aortitis. Wnk1 deletion in VSMCs led to aortic wall remodeling with destruction of elastin layers, increased collagen content, and enhanced local TGF-β (transforming growth factor-beta) 1 expression. Finally, in vivo TGF-β blockade using neutralizing anti-TGF-β antibody promoted saccular aneurysm formation and aorta rupture in Sm22 Cre+ Wnk1lox/lox mice but not in control animals. CONCLUSION Wnk1 is a key regulator of VSMC function. Wnk1 deletion promotes VSMC phenotype switch toward a pathogenic proinflammatory phenotype, orchestrating deleterious vascular remodeling and spontaneous severe aortitis in mice.
Collapse
MESH Headings
- Animals
- Aortitis/genetics
- Aortitis/metabolism
- Aortitis/pathology
- Mice
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Angiotensin II
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Humans
- WNK Lysine-Deficient Protein Kinase 1/genetics
- WNK Lysine-Deficient Protein Kinase 1/metabolism
- Mice, Inbred C57BL
- Male
- Cells, Cultured
- Mice, Knockout, ApoE
- Disease Models, Animal
- Inflammation/metabolism
- Inflammation/genetics
- Inflammation/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
Collapse
Affiliation(s)
- Helene Quelquejay
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Rida Al-Rifai
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Michele Silvestro
- Division of Vascular and Endovascular Surgery, Department of Surgery and Department of Cell Biology, New York University Langone Medical Center (M.S., B.R.)
| | - Marie Vandestienne
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Irmine Ferreira
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Tristan Mirault
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Daniel Henrion
- MITOVASC Department, Team 2 (CarMe), ICAT SFR (Interactions Cellulaires et Applications Thérapeutiques Structure Fédérale de Recherche), University of Angers, Inserm U1083, France (D.H.)
| | - Xiaodan Zhong
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Icia Santos-Zas
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
- Laboratorio de Endocrinología Celular, Área de Endocrinología Molecular y Celular Instituto de Investigación Sanitaria de Santiago, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain (I.S.-Z.)
| | - Guillaume Goudot
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Paul Alayrac
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Estelle Robidel
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Gwennhael Autret
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Daniel Balvay
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Soraya Taleb
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Alain Tedgui
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Chantal M Boulanger
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (A.Z., C.C.)
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-Based Infection Research, Helmholtz-Center for Infection Research, Würzburg, Germany (A.-E.S.)
| | - Juliette Hadchouel
- Inserm UMRS 1155, Tenon Hospital (J.H.), Sorbonne Université, Paris, France
| | - Bhama Ramkhelawon
- Division of Vascular and Endovascular Surgery, Department of Surgery and Department of Cell Biology, New York University Langone Medical Center (M.S., B.R.)
| | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (A.Z., C.C.)
| | - Sonia Bergaya
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Xavier Jeunemaitre
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Hafid Ait-Oufella
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP (Assistance Publique- Hôpitaux de Paris) (H.A.-O.), Sorbonne Université, Paris, France
| |
Collapse
|
8
|
Krebs JR, Bellotti P, Valisno JAC, Su G, Sharma S, Kollareth DJM, Hartman JB, Adithan A, Spinosa M, Kamat M, Garrett T, Cai G, Sharma AK, Upchurch GR. Pharmacologic Inhibition of Ferroptosis Attenuates Experimental Abdominal Aortic Aneurysm Formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599427. [PMID: 39149340 PMCID: PMC11326422 DOI: 10.1101/2024.06.18.599427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) formation involves vascular inflammation, thrombosis formation and programmed cell death leading to aortic remodeling. Recent studies have suggested that ferroptosis, an excessive iron-mediated cell death, can regulate cardiovascular diseases, including AAAs. However, the role of ferroptosis in immune cells, like macrophages, and ferroptosis-related genes in AAA formation remains to be deciphered. Single cell-RNA sequencing of human aortic tissue from AAA patients demonstrates significant differences in ferroptosis-related genes compared to control aortic tissue. Using two established murine models of AAA and aortic rupture in C57BL/6 (WT) mice, we observed that treatment with liproxstatin-1, a specific ferroptosis inhibitor, significantly attenuated aortic diameter, pro-inflammatory cytokine production, immune cell infiltration (neutrophils and macrophages), increased smooth muscle cell α-actin expression and elastic fiber disruption compared to mice treated with inactivated elastase in both pre-treatment and treatment after a small AAA had already formed. Lipidomic analysis using mass spectrometry shows a significant increase in ceramides and a decrease in intact lipid species levels in murine tissue compared to controls in the chronic AAA model on day 28. Mechanistically, in vitro studies demonstrate that liproxstatin-1 treatment of macrophages mitigated the crosstalk with aortic smooth muscle cells (SMCs) by downregulating MMP2 secretion. Taken together, this study demonstrates that pharmacological inhibition by liproxstatin-1 mitigates macrophage-dependent ferroptosis contributing to inhibition of aortic inflammation and remodeling during AAA formation.
Collapse
|
9
|
Chu S, Shan D, He L, Yang S, Feng Y, Zhang Y, Yu J. Anemoside B4 attenuates abdominal aortic aneurysm by limiting smooth muscle cell transdifferentiation and its mediated inflammation. Front Immunol 2024; 15:1412022. [PMID: 38881898 PMCID: PMC11176519 DOI: 10.3389/fimmu.2024.1412022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative disease characterized by local abnormal dilation of the aorta accompanied by vascular smooth muscle cell (VSMC) dysfunction and chronic inflammation. VSMC dedifferentiation, transdifferentiation, and increased expression of matrix metalloproteinases (MMPs) are essential causes of AAA formation. Previous studies from us and others have shown that Anemoside B4 (AB4), a saponin from Pulsatilla chinensis, has anti-inflammatory, anti-tumor, and regulatory effects on VSMC dedifferentiation. The current study aimed to investigate whether AB4 inhibits AAA development and its underlying mechanisms. By using an Ang II induced AAA model in vivo and cholesterol loading mediated VSMC to macrophage transdifferentiation model in vitro, our study demonstrated that AB4 could attenuate AAA pathogenesis, prevent VSMC dedifferentiation and transdifferentiation to macrophage-like cells, decrease vascular inflammation, and suppress MMP expression and activity. Furthermore, KLF4 overexpression attenuated the effects of AB4 on VSMC to macrophage-like cell transition and VSMC inflammation in vitro. In conclusion, AB4 protects against AAA formation in mice by inhibiting KLF4 mediated VSMC transdifferentiation and inflammation. Our study provides the first proof of concept of using AB4 for AAA management.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/chemically induced
- Cell Transdifferentiation/drug effects
- Kruppel-Like Factor 4/metabolism
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Inflammation/metabolism
- Saponins/pharmacology
- Disease Models, Animal
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Mice, Inbred C57BL
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/immunology
- Angiotensin II/pharmacology
- Humans
Collapse
Affiliation(s)
- Shuhan Chu
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Dan Shan
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Luling He
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Shilin Yang
- National Pharmaceutical Engineering Center (NPEC) for Solid Preparation in Chinese Herbal Medicine, Nanchang, Jiangxi, China
| | - Yulin Feng
- National Pharmaceutical Engineering Center (NPEC) for Solid Preparation in Chinese Herbal Medicine, Nanchang, Jiangxi, China
| | - Yifeng Zhang
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
10
|
Wei B, Deng N, Guo H, Wei Y, Xu F, Luo S, You W, Chen J, Li W, Si X. Trimethylamine N-oxide promotes abdominal aortic aneurysm by inducing vascular inflammation and vascular smooth muscle cell phenotypic switching. Eur J Pharmacol 2024; 965:176307. [PMID: 38160930 DOI: 10.1016/j.ejphar.2023.176307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 12/04/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE Inflammation and vascular smooth muscle cell (VSMC) phenotypic switching are implicated in the pathogenesis of abdominal aortic aneurysm (AAA). Trimethylamine N-oxide (TMAO) has emerged as a crucial risk factor in cardiovascular diseases, inducing vascular inflammation and calcification. We aimed to evaluate the effect of TMAO on the formation of AAA. APPROACH AND RESULTS Here, we showed that TMAO was elevated in plasma from AAA patients compared with nonaneurysmal subjects by liquid chromatography‒mass spectrometry (LC‒MS) detection. Functional studies revealed that increased TMAO induced by feeding a choline-supplemented diet promoted Ang II-induced AAA formation. Immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), and Western blot analyses revealed that TMAO induced macrophage infiltration and inflammatory factor release. Conversely, inhibition of TMAO by supplementation with DMB suppressed AAA formation and the inflammatory response. Molecular studies revealed that TMAO regulated VSMC phenotypic switching. Flow cytometry analyses showed that TMAO induces macrophage M1-type polarization. Furthermore, pharmacological intervention experiments suggested that the nuclear factor-κB (NF-κB) signaling pathway was critical for TMAO to trigger AAA formation. CONCLUSIONS TMAO promotes AAA formation by inducing vascular inflammation and VSMC phenotypic switching through activation of the NF-κB signaling pathway. Thus, TMAO is a prospective therapeutic AAA target.
Collapse
Affiliation(s)
- Bo Wei
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China
| | - Na Deng
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China
| | - Haijun Guo
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China
| | - Yingying Wei
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China
| | - Fujia Xu
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China
| | - Sihan Luo
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China
| | - Weili You
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China
| | - Jingjing Chen
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China
| | - Wei Li
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China.
| | - Xiaoyun Si
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang, 550004, Guizhou Province, China.
| |
Collapse
|
11
|
Dubner AM, Lu S, Jolly AJ, Strand KA, Mutryn MF, Hinthorn T, Noble T, Nemenoff RA, Moulton KS, Majesky MW, Weiser-Evans MC. Smooth muscle-derived adventitial progenitor cells direct atherosclerotic plaque composition complexity in a Klf4-dependent manner. JCI Insight 2023; 8:e174639. [PMID: 37991018 PMCID: PMC10755692 DOI: 10.1172/jci.insight.174639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/05/2023] [Indexed: 11/23/2023] Open
Abstract
We previously established that vascular smooth muscle-derived adventitial progenitor cells (AdvSca1-SM) preferentially differentiate into myofibroblasts and contribute to fibrosis in response to acute vascular injury. However, the role of these progenitor cells in chronic atherosclerosis has not been defined. Using an AdvSca1-SM cell lineage tracing model, scRNA-Seq, flow cytometry, and histological approaches, we confirmed that AdvSca1-SM-derived cells localized throughout the vessel wall and atherosclerotic plaques, where they primarily differentiated into fibroblasts, smooth muscle cells (SMC), or remained in a stem-like state. Krüppel-like factor 4 (Klf4) knockout specifically in AdvSca1-SM cells induced transition to a more collagen-enriched fibroblast phenotype compared with WT mice. Additionally, Klf4 deletion drastically modified the phenotypes of non-AdvSca1-SM-derived cells, resulting in more contractile SMC and atheroprotective macrophages. Functionally, overall plaque burden was not altered with Klf4 deletion, but multiple indices of plaque composition complexity, including necrotic core area, macrophage accumulation, and fibrous cap thickness, were reduced. Collectively, these data support that modulation of AdvSca1-SM cells through KLF4 depletion confers increased protection from the development of potentially unstable atherosclerotic plaques.
Collapse
Affiliation(s)
- Allison M. Dubner
- Department of Medicine, Division of Renal Diseases and Hypertension
- Integrated Physiology PhD Program
| | - Sizhao Lu
- Department of Medicine, Division of Renal Diseases and Hypertension
- School of Medicine, Consortium for Fibrosis Research and Translation
| | - Austin J. Jolly
- Department of Medicine, Division of Renal Diseases and Hypertension
- Medical Scientist Training Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Keith A. Strand
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Marie F. Mutryn
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Tyler Hinthorn
- Department of Medicine, Division of Renal Diseases and Hypertension
- Biomedical Sciences and Biotechnology MS program, University of Colorado Graduate School, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tysen Noble
- Department of Medicine, Division of Renal Diseases and Hypertension
- Biomedical Sciences and Biotechnology MS program, University of Colorado Graduate School, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Raphael A. Nemenoff
- Department of Medicine, Division of Renal Diseases and Hypertension
- School of Medicine, Consortium for Fibrosis Research and Translation
| | - Karen S. Moulton
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark W. Majesky
- Center for Developmental Biology & Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, USA
- Departments of Pediatrics, Laboratory Medicine & and Pathology, University of Washington, Seattle, Washington, USA
| | - Mary C.M. Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension
- Integrated Physiology PhD Program
- School of Medicine, Consortium for Fibrosis Research and Translation
- Medical Scientist Training Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
- Cardiovascular Pulmonary Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
12
|
Bharadhwaj RA, Kumarswamy R. Long noncoding RNA TUG1 regulates smooth muscle cell differentiation via KLF4-myocardin axis. Am J Physiol Cell Physiol 2023; 325:C940-C950. [PMID: 37642238 PMCID: PMC10635660 DOI: 10.1152/ajpcell.00275.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Abdominal aortic aneurysms (AAAs) are asymptomatic vascular diseases that have life-threatening outcomes. Smooth muscle cell (SMC) dysfunction plays an important role in AAA development. The contribution of non-coding genome, specifically the role of long non-coding RNAs (lncRNAs) in SMC dysfunction, is relatively unexplored. We investigated the role of lncRNA TUG1 in SMC dysfunction. To identify potential lncRNAs relevant to SMC functionality, lncRNA profiling was performed in angiotensin-II-treated SMCs. AAA was induced by angiotensin-II treatment in mice. Transcriptional regulation of TUG1 was studied using promoter luciferase and chromatin-immuno-precipitation experiments. Gain-or-loss-of-function experiments were performed in vitro to investigate TUG1-mediated regulation of SMC function. Immunoprecipitation experiments were conducted to elucidate the mechanism underlying TUG1-mediated SMC dysfunction. TUG1 was upregulated in SMCs following angiotensin-II treatment. Similarly, TUG1 levels were elevated in abdominal aorta in a mouse model of angiotensin-II-induced AAA. Further investigations showed that angiotensin-II-induced TUG1 expression could be suppressed by inhibiting Notch-signaling pathway, both in vitro and in mouse AAA model and that TUG1 is a direct transcriptional target of the Notch pathway. In aneurysmal tissues, TUG1 expression was inversely correlated with the expression of SMC contractile genes. Overexpression of TUG1 repressed SMC differentiation in vitro, whereas siRNA/shRNA-mediated TUG1 knockdown showed an opposite effect. Mechanistically, TUG1 interacts with transcriptional repressor KLF4 and facilitates its recruitment to myocardin promoter ultimately leading to the repression of SMC differentiation. In summary, our study uncovers a novel role for the lncRNA TUG1 wherein it modulates SMC differentiation via the KLF4-myocardin axis, which may have potential implications in AAA development.NEW & NOTEWORTHY TUG1 is an angiotensin-II-induced long noncoding RNA that mediates smooth muscle cell (SMC) dysfunction through interaction with transcriptional repressor KLF4.
Collapse
Affiliation(s)
- Ravi Abishek Bharadhwaj
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Regalla Kumarswamy
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
13
|
Lei C, Kan H, Xian X, Chen W, Xiang W, Song X, Wu J, Yang D, Zheng Y. FAM3A reshapes VSMC fate specification in abdominal aortic aneurysm by regulating KLF4 ubiquitination. Nat Commun 2023; 14:5360. [PMID: 37660071 PMCID: PMC10475135 DOI: 10.1038/s41467-023-41177-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 08/24/2023] [Indexed: 09/04/2023] Open
Abstract
Reprogramming of vascular smooth muscle cell (VSMC) differentiation plays an essential role in abdominal aortic aneurysm (AAA). However, the underlying mechanisms are still unclear. We explore the expression of FAM3A, a newly identified metabolic cytokine, and whether and how FAM3A regulates VSMC differentiation in AAA. We discover that FAM3A is decreased in the aortas and plasma in AAA patients and murine models. Overexpression or supplementation of FAM3A significantly attenuate the AAA formation, manifested by maintenance of the well-differentiated VSMC status and inhibition of VSMC transformation toward macrophage-, chondrocyte-, osteogenic-, mesenchymal-, and fibroblast-like cell subpopulations. Importantly, FAM3A induces KLF4 ubiquitination and reduces its phosphorylation and nuclear localization. Here, we report FAM3A as a VSMC fate-shaping regulator in AAA and reveal the underlying mechanism associated with KLF4 ubiquitination and stability, which may lead to the development of strategies based on FAM3A to restore VSMC homeostasis in AAA.
Collapse
Affiliation(s)
- Chuxiang Lei
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Haoxuan Kan
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Xiangyu Xian
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Wenlin Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Wenxuan Xiang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Xiaohong Song
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Jianqiang Wu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Dan Yang
- Department of Computational Biology and Bioinformatics, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Haidian District, Beijing, 100193, China.
| | - Yuehong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
14
|
Hu Y, Cai Z, He B. Smooth Muscle Heterogeneity and Plasticity in Health and Aortic Aneurysmal Disease. Int J Mol Sci 2023; 24:11701. [PMID: 37511460 PMCID: PMC10380637 DOI: 10.3390/ijms241411701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the medial layer of the aorta, which plays a critical role in the maintenance of aortic wall integrity. VSMCs have been suggested to have contractile and synthetic phenotypes and undergo phenotypic switching to contribute to the deteriorating aortic wall structure. Recently, the unprecedented heterogeneity and diversity of VSMCs and their complex relationship to aortic aneurysms (AAs) have been revealed by high-resolution research methods, such as lineage tracing and single-cell RNA sequencing. The aortic wall consists of VSMCs from different embryonic origins that respond unevenly to genetic defects that directly or indirectly regulate VSMC contractile phenotype. This difference predisposes to hereditary AAs in the aortic root and ascending aorta. Several VSMC phenotypes with different functions, for example, secreting VSMCs, proliferative VSMCs, mesenchymal stem cell-like VSMCs, immune-related VSMCs, proinflammatory VSMCs, senescent VSMCs, and stressed VSMCs are identified in non-hereditary AAs. The transformation of VSMCs into different phenotypes is an adaptive response to deleterious stimuli but can also trigger pathological remodeling that exacerbates the pathogenesis and development of AAs. This review is intended to contribute to the understanding of VSMC diversity in health and aneurysmal diseases. Papers that give an update on VSMC phenotype diversity in health and aneurysmal disease are summarized and recent insights on the role of VSMCs in AAs are discussed.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
15
|
Dubner AM, Lu S, Jolly AJ, Strand KA, Mutryn MF, Hinthorn T, Noble T, Nemenoff RA, Moulton KS, Majesky MW, Weiser-Evans MCM. Smooth muscle-derived adventitial progenitor cells promote key cell type transitions controlling plaque stability in atherosclerosis in a Klf4-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.18.549539. [PMID: 37503181 PMCID: PMC10370085 DOI: 10.1101/2023.07.18.549539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
We previously established that vascular smooth muscle-derived adventitial progenitor cells (AdvSca1-SM) preferentially differentiate into myofibroblasts and contribute to fibrosis in response to acute vascular injury. However, the role of these progenitor cells in chronic atherosclerosis has not been defined. Using an AdvSca1-SM lineage tracing model, scRNA-Seq, flow cytometry, and histological approaches, we confirmed that AdvSca1-SM cells localize throughout the vessel wall and atherosclerotic plaques, where they primarily differentiate into fibroblasts, SMCs, or remain in a stem-like state. Klf4 knockout specifically in AdvSca1-SM cells induced transition to a more collagen-enriched myofibroblast phenotype compared to WT mice. Additionally, Klf4 depletion drastically modified the phenotypes of non-AdvSca1-SM-derived cells, resulting in more contractile SMCs and atheroprotective macrophages. Functionally, overall plaque burden was not altered with Klf4 depletion, but multiple indices of plaque vulnerability were reduced. Collectively, these data support that modulating the AdvSca1-SM population confers increased protection from the development of unstable atherosclerotic plaques.
Collapse
Affiliation(s)
- Allison M Dubner
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Integrated Physiology PhD Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Sizhao Lu
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Austin J Jolly
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Keith A Strand
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marie F Mutryn
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tyler Hinthorn
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Biomedical Sciences and Biotechnology MS program, University of Colorado Graduate School, Anschutz Medical Campus, Aurora, CO, USA
| | - Tysen Noble
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Biomedical Sciences and Biotechnology MS program, University of Colorado Graduate School, Anschutz Medical Campus, Aurora, CO, USA
| | - Raphael A Nemenoff
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Karen S Moulton
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mark W Majesky
- Center for Developmental Biology & Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA 98101
- Departments of Pediatrics, Laboratory Medicine & and Pathology, University of Washington, Seattle, WA, 98195
| | - Mary CM Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Integrated Physiology PhD Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cardiovascular Pulmonary Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
16
|
Wang Y, Liu X, Xu Q, Xu W, Zhou X, Lin Z. CCN2 deficiency in smooth muscle cells triggers cell reprogramming and aggravates aneurysm development. JCI Insight 2023; 8:162987. [PMID: 36625347 PMCID: PMC9870081 DOI: 10.1172/jci.insight.162987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/17/2022] [Indexed: 01/11/2023] Open
Abstract
Vascular smooth muscle cell (SMC) phenotypic switching is widely recognized as a key mechanism responsible for the pathogenesis of several aortic diseases, such as aortic aneurysm. Cellular communication network factor 2 (CCN2), often upregulated in human pathologies and animal disease models, exerts myriad context-dependent biological functions. However, current understanding of the role of SMC-CCN2 in SMC phenotypic switching and its function in the pathology of abdominal aortic aneurysm (AAA) is lacking. Here, we show that SMC-restricted CCN2 deficiency causes AAA in the infrarenal aorta of angiotensin II-infused (Ang II-infused) hypercholesterolemic mice at a similar anatomic location to human AAA. Notably, the resistance of naive C57BL/6 WT mice to Ang II-induced AAA formation is lost upon silencing of CCN2 in SMC. Furthermore, the pro-AAA phenotype of SMC-CCN2-KO mice is recapitulated in a different model that involves the application of elastase-β-aminopropionitrile. Mechanistically, our findings reveal that CCN2 intersects with TGF-β signaling and regulates SMC marker expression. Deficiency of CCN2 triggers SMC reprograming associated with alterations in Krüppel-like factor 4 and contractile marker expression, and this reprograming likely contributes to the development of AAA in mice. These results identify SMC-CCN2 as potentially a novel regulator of SMC phenotypic switching and AA biology.
Collapse
Affiliation(s)
- Yu Wang
- Cardiology Division, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Xuesong Liu
- Cardiology Division, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qian Xu
- Cardiology Division, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Wei Xu
- Cardiology Division, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Xianming Zhou
- Cardiology Division, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Lin
- Cardiology Division, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Lai PMR, Ryu JY, Park SC, Gross BA, Dickinson LD, Dagen S, Aziz-Sultan MA, Boulos AS, Barrow DL, Batjer HH, Blackburn S, Chang EF, Chen PR, Colby GP, Cosgrove GR, David CA, Day AL, Frerichs KU, Niemela M, Ojemann SG, Patel NJ, Shi X, Valle-Giler EP, Wang AC, Welch BG, Zusman EE, Weiss ST, Du R. Somatic Variants in SVIL in Cerebral Aneurysms. Neurol Genet 2022; 8:e200040. [PMID: 36475054 PMCID: PMC9720733 DOI: 10.1212/nxg.0000000000200040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
Background and ObjectivesWhile somatic mutations have been well-studied in cancer, their roles in other complex traits are much less understood. Our goal is to identify somatic variants that may contribute to the formation of saccular cerebral aneurysms.MethodsWe performed whole-exome sequencing on aneurysm tissues and paired peripheral blood. RNA sequencing and the CRISPR/Cas9 system were then used to perform functional validation of our results.ResultsSomatic variants involved in supervillin (SVIL) or its regulation were found in 17% of aneurysm tissues. In the presence of a mutation in theSVILgene, the expression level of SVIL was downregulated in the aneurysm tissue compared with normal control vessels. Downstream signaling pathways that were induced by knockdown ofSVILvia the CRISPR/Cas9 system in vascular smooth muscle cells (vSMCs) were determined by evaluating changes in gene expression and protein kinase phosphorylation. We found thatSVILregulated the phenotypic modulation of vSMCs to the synthetic phenotype via Krüppel-like factor 4 and platelet-derived growth factor and affected cell migration of vSMCs via the RhoA/ROCK pathway.DiscussionWe propose that somatic variants form a novel mechanism for the development of cerebral aneurysms. Specifically, somatic variants inSVILresult in the phenotypic modulation of vSMCs, which increases the susceptibility to aneurysm formation. This finding suggests a new avenue for the therapeutic intervention and prevention of cerebral aneurysms.
Collapse
Affiliation(s)
- Pui Man Rosalind Lai
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jee-Yeon Ryu
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sang-Cheol Park
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Bradley A Gross
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lawrence D Dickinson
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sarajune Dagen
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mohammad Ali Aziz-Sultan
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Alan S Boulos
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Daniel L Barrow
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - H Hunt Batjer
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Spiros Blackburn
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Edward F Chang
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - P Roc Chen
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Geoffrey P Colby
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Garth Rees Cosgrove
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Carlos A David
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Arthur L Day
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kai U Frerichs
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mika Niemela
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Steven G Ojemann
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Nirav J Patel
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Xiangen Shi
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Edison P Valle-Giler
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Anthony C Wang
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Babu G Welch
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Edie E Zusman
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Scott T Weiss
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rose Du
- Department of Neurosurgery (P.M.R.L., J.-Y.R., S.-C.P., S.D., M.A.A.-S., G.R.C., K.U.F., N.J.P., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Artificial Intelligence and Robotics Laboratory (S.-C.P.), Myongji Hospital, Goyang, Korea; Department of Neurosurgery (B.A.G.), University of Pittsburgh, PA; Department of Neurosurgery (L.D.D., E.E.Z.), Sutter Health, Danville, CA; Department of Neurosurgery (A.S.B.), Albany Medical Center, NY; Department of Neurosurgery (D.L.B.), Emory University, Atlanta, GA; Department of Neurosurgery (H.H.B., B.G.W.), University of Texas Southwestern, Dallas, TX; Department of Neurosurgery (S.B., P.R.C., A.L.D.), University of Texas Health Science Center, Houston; Department of Neurosurgery (E.F.C.), University of California San Francisco, CA; Department of Neurosurgery (G.P.C., A.C.W.), University of California Los Angeles; Department of Neurosurgery (C.A.D.), Lahey Hospital and Medical Center, Burlington, MA; Department of Neurosurgery (M.N.), Helsinki University and Helsinki University Hospital, Finland; Department of Neurosurgery (S.G.O.), University of Colorado, Denver; Department of Neurosurgery (X.S.), Affiliated Fuxing Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery (E.P.V.-G.), Ochsner Medical Center, New Orleans, LA; and Channing Division of Network Medicine (S.T.W., R.D.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Sun LY, Lyu YY, Zhang HY, Shen Z, Lin GQ, Geng N, Wang YL, Huang L, Feng ZH, Guo X, Lin N, Ding S, Yuan AC, Zhang L, Qian K, Pu J. Nuclear Receptor NR1D1 Regulates Abdominal Aortic Aneurysm Development by Targeting the Mitochondrial Tricarboxylic Acid Cycle Enzyme Aconitase-2. Circulation 2022; 146:1591-1609. [PMID: 35880522 PMCID: PMC9674448 DOI: 10.1161/circulationaha.121.057623] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 06/10/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Metabolic disorder increases the risk of abdominal aortic aneurysm (AAA). NRs (nuclear receptors) have been increasingly recognized as important regulators of cell metabolism. However, the role of NRs in AAA development remains largely unknown. METHODS We analyzed the expression profile of the NR superfamily in AAA tissues and identified NR1D1 (NR subfamily 1 group D member 1) as the most highly upregulated NR in AAA tissues. To examine the role of NR1D1 in AAA formation, we used vascular smooth muscle cell (VSMC)-specific, endothelial cell-specific, and myeloid cell-specific conditional Nr1d1 knockout mice in both AngII (angiotensin II)- and CaPO4-induced AAA models. RESULTS Nr1d1 gene expression exhibited the highest fold change among all 49 NRs in AAA tissues, and NR1D1 protein was upregulated in both human and murine VSMCs from AAA tissues. The knockout of Nr1d1 in VSMCs but not endothelial cells and myeloid cells inhibited AAA formation in both AngII- and CaPO4-induced AAA models. Mechanistic studies identified ACO2 (aconitase-2), a key enzyme of the mitochondrial tricarboxylic acid cycle, as a direct target trans-repressed by NR1D1 that mediated the regulatory effects of NR1D1 on mitochondrial metabolism. NR1D1 deficiency restored the ACO2 dysregulation and mitochondrial dysfunction at the early stage of AngII infusion before AAA formation. Supplementation with αKG (α-ketoglutarate, a downstream metabolite of ACO2) was beneficial in preventing and treating AAA in mice in a manner that required NR1D1 in VSMCs. CONCLUSIONS Our data define a previously unrecognized role of nuclear receptor NR1D1 in AAA pathogenesis and an undescribed NR1D1-ACO2 axis involved in regulating mitochondrial metabolism in VSMCs. It is important that our findings suggest αKG supplementation as an effective therapeutic approach for AAA treatment.
Collapse
MESH Headings
- Humans
- Mice
- Animals
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/prevention & control
- Aorta, Abdominal/pathology
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- Muscle, Smooth, Vascular/metabolism
- Citric Acid Cycle
- Myocytes, Smooth Muscle/metabolism
- Angiotensin II/adverse effects
- Mice, Knockout
- Aconitate Hydratase/metabolism
- Disease Models, Animal
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Ling-Yue Sun
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Yan Lyu
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Heng-Yuan Zhang
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Shen
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Guan-Qiao Lin
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Na Geng
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Li Wang
- Department of Vascular Surgery (Y.-L.W., L.Z.), Shanghai Jiao Tong University, Shanghai, China
| | - Lin Huang
- Renji Hospital, School of Medicine, School of Biomedical Engineering and Med-X Research Institute (L.H., K.Q.), Shanghai Jiao Tong University, Shanghai, China
| | - Ze-Hao Feng
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Guo
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Nan Lin
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Song Ding
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - An-Cai Yuan
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Lan Zhang
- Department of Vascular Surgery (Y.-L.W., L.Z.), Shanghai Jiao Tong University, Shanghai, China
| | - Kun Qian
- Renji Hospital, School of Medicine, School of Biomedical Engineering and Med-X Research Institute (L.H., K.Q.), Shanghai Jiao Tong University, Shanghai, China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Zhou X, Liu G, Lai H, Wang C, Li J, Zhu K. Using Molecular Targets to Predict and Treat Aortic Aneurysms. Rev Cardiovasc Med 2022; 23:307. [PMID: 39077712 PMCID: PMC11262374 DOI: 10.31083/j.rcm2309307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 07/31/2024] Open
Abstract
Aortic aneurysms are life-threatening vascular diseases associated with high morbidity, and usually require prophylactic surgical intervention. Current preventative management of aortic aneurysms relies on the diameter and other anatomic parameters of the aorta, but these have been demonstrated to be insufficient predictive factors of disease progression and potential complications. Studies on pathophysiology of aortic aneurysms could fill this need, which already indicated the significance of specific molecules in aortic aneurysms. These molecules provide more accurate prediction, and they also serve as therapeutic targets, some of which are in preclinical stage. In this review, we summarized the inadequacies and achievements of current clinical prediction standards, discussed the molecular targets in prediction and treatment, and especially emphasized the molecules that have shown potentials in early diagnosis, accurate risk assessment and target treatment of aortic aneurysm at early stage.
Collapse
Affiliation(s)
- Xiaonan Zhou
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Gang Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| |
Collapse
|
20
|
Xu H, Yu M, Yu Y, Li Y, Yang F, Liu Y, Han L, Xu Z, Wang G. KLF4 prevented angiotensin II-induced smooth muscle cell senescence by enhancing autophagic activity. Eur J Clin Invest 2022; 52:e13804. [PMID: 35506324 DOI: 10.1111/eci.13804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Vascular aging is an important risk factor for various cardiovascular diseases. Transcription factor krüppel-like factor 4 (KLF4) could regulate the phenotypic transformation of the vascular smooth muscle cell (VSMC) in the pathogenesis of aortic diseases. The present study aimed to explore the role and mechanism of KLF4 in angiotensin II (Ang II)-induced VSMC senescence. METHODS The VSMC senescence mouse model was induced by sustained release of Ang II (1.0 μg/kg/min) for 4 weeks. The premature senescent VSMCs were induced by Ang II (0.1 μmol/L) for 72 h. Cellular senescence was measured by senescence-associated β-galactosidase (SA-β-gal) activity and p53/p16 expression. The autophagic activity was evaluated by autophagic flux and autophagic marker expression. RESULTS The expression of KLF4 was extremely increased in abdominal aorta tissues after 1-week Ang II stimulation (p < .01) but began to decrease in later periods. Decreased expression of KLF4 was also detected in premature senescent VSMCs. Overexpression of KLF4 could enhance the antisenescence ability of VSMCs. Significantly decreased amounts of SA-β-gal-positive cells and lower p53/p16 expression were detected in KLF4-overexpressing VSMCs (p < .01). Next, telomerase reverse transcriptase (TERT) was identified as a direct downstream target of KLF4 in VSMCs. Overexpression of KLF4 in VSMCs prevented the decreased expression of TERT under Ang II stimulation condition, which could in turn, contribute to the enhanced autophagic activity, and ultimately to the improved antisenescence ability of VSMCs. CONCLUSIONS Our results demonstrated that overexpression of KLF4 prevented Ang II-induced VSMC senescence by promoting TERT-mediated autophagy. These findings provided novel potential targets for the prevention and therapy of vascular aging.
Collapse
Affiliation(s)
- Hongjie Xu
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Manli Yu
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yongchao Yu
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yang Li
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Fan Yang
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yang Liu
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China.,Department of Critical Care Medicine, Naval Medical Center of PLA, Shanghai, China
| | - Lin Han
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guokun Wang
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
21
|
Salmon M, Hawkins RB, Dahl J, Scott E, Johnston WF, Ailawadi G. Genetic and Pharmacological Disruption of Interleukin-1α Leads to Augmented Murine Aortic Aneurysm. Ann Vasc Surg 2022; 85:358-370. [PMID: 35680012 PMCID: PMC11029039 DOI: 10.1016/j.avsg.2022.05.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/28/2022] [Accepted: 05/11/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND Interleukin-1 (IL-1) signaling has an established role as a cytokine signaling pathway important for progression of abdominal aortic aneurysms (AAAs). While the IL-1β ligand and IL-1R1 have been previously investigated, the role of the IL-1α ligand in AAAs remains unknown. In this study, we sought to examine the role of IL-1α in AAAs using genetic and pharmacologic approaches. METHODS Eight-week-old wild-type (WT) or IL-1α knock-out (KO) male and female mice (n = 10-16/group) underwent experimental AAA and were harvested 14 days following surgery to assess AAA size and characteristics. In separate studies, 8-week-old WT mice were treated with an inhibitor to IL-1α during AAA formation and harvested 14 days following surgery. Finally, WT and IL-1α KO mice were administered Anakinra, an IL-R1 inhibitor, during AAA formation to determine the effect of inhibiting IL-1R1 when IL-1α is knocked out. RESULTS Male and female IL-1α KO mice had larger AAAs compared to WT AAAs (male: 153% vs. 89.2%, P = 0.0001; female: 86.6% vs. 63.5%, P = 0.02). IL-1α KO mice had greater elastin breakage (P = 0.01), increased levels of macrophage staining (P = 0.0045), and greater pro-metallo proteinase 2 (P = 0.02). Pharmacologic inhibition of WT male mice with an IL-1α neutralizing antibody resulted in larger AAAs (133.1% vs. 77.0%, P < 0.001). Finally, treatment of IL-1α KO male mice with Anakinra decreased AAA formation compared with vehicle control AAAs (Anakinra + IL-1α KO: 47.7% vs. WT: 147.1%; P = 0.0001). CONCLUSIONS IL-1α disruption using either genetic or pharmacologic approaches worsens AAAs.
Collapse
Affiliation(s)
- Morgan Salmon
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA; Department of Cardiac Surgery, University of Michigan School of Medicine, Ann Arbor, MI; Frankel Cardiovascular Center, University of Michigan School of Medicine, Ann Arbor, MI; Department of Surgery, Oschner Medical Center, New Orleans, LA.
| | - Robert B Hawkins
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA; Department of Cardiac Surgery, University of Michigan School of Medicine, Ann Arbor, MI; Frankel Cardiovascular Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Jolian Dahl
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA
| | - Erik Scott
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA
| | | | - Gorav Ailawadi
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA; Department of Cardiac Surgery, University of Michigan School of Medicine, Ann Arbor, MI; Frankel Cardiovascular Center, University of Michigan School of Medicine, Ann Arbor, MI
| |
Collapse
|
22
|
Lei L, Zhou Y, Wang T, Zheng Z, Chen L, Pan Y. Activation of AMP-activated protein kinase ablated the formation of aortic dissection by suppressing vascular inflammation and phenotypic switching of vascular smooth muscle cells. Int Immunopharmacol 2022; 112:109177. [PMID: 36049351 DOI: 10.1016/j.intimp.2022.109177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/08/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Aortic dissection (AD) is a fatal vascular disease in absence of effective pharmaceutical therapy. Adenosine monophosphate-activated protein kinase α (AMPKα) plays a critical role in various cardiovascular diseases. Whether AMPKα is involved in the pathogenesis of aortic dissection remains unknown. We aimed to determine whether activation of AMPKα prevents the formation of AD. METHODS AND RESULTS Reduced expression of phosphorylated AMPKα (Thr172) and exacerbated phenotypic switching were observed in human aortic tissues from aortic dissection patients compared with those in tissues from controls. In vivo, the formation of aortic dissection in ApoE-/- mice was successfully induced by continuous infusion of angiotensin II (AngII) for two weeks, characterized by the activation of vascular inflammation, infiltration of macrophages and phenotypic switching of vascular smooth muscle cells (VSMCs). rAAV2-mediated overexpression of constitutively active AMPKα (CA-AMPKα) enhanced the expression of phosphorylated AMPKα (Thr172) and attenuated AngII-induced occurrence of aortic dissection by suppressing the infiltration of macrophages, activation of vascular inflammation and phenotypic switching of VSMCs. The pathogenesis above was conversely exacerbated by rAAV2-mediated overexpression of dominant negative AMPKα2 (DN-AMPKα). In vitro, we demonstrated that the administration of an AMPK agonist (AICAR) or transfection of CA-AMPKα induced the activation of AMPKα and then ameliorated AngII-induced phenotypic switching in the VSMCs and inflammation in the bone marrow-derived macrophages (BMDMs). This could be reversed by the addition of AMPK inhibitor compound C or transfection of DN-AMPKα. CONCLUSION Impaired activation of AMPKα may increase the susceptibility to aortic dissection. Our findings verified the protective effects of AMPKα on the formation of aortic dissection and may provide evidence for clinical prevention or treatment.
Collapse
Affiliation(s)
- Lei Lei
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanrong Zhou
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tiemao Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhi Zheng
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang Chen
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Youmin Pan
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
23
|
Van Hoose PM, Yang L, Kraemer M, Ubele M, Morris AJ, Smyth SS. Lipid phosphate phosphatase 3 in smooth muscle cells regulates angiotensin II-induced abdominal aortic aneurysm formation. Sci Rep 2022; 12:5664. [PMID: 35383201 PMCID: PMC8983654 DOI: 10.1038/s41598-022-08422-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/10/2021] [Indexed: 01/28/2023] Open
Abstract
Genetic variants that regulate lipid phosphate phosphatase 3 (LPP3) expression are risk factors for the development of atherosclerotic cardiovascular disease. LPP3 is dynamically upregulated in the context of vascular inflammation with particularly heightened expression in smooth muscle cells (SMC), however, the impact of LPP3 on vascular pathology is not fully understood. We investigated the role of LPP3 and lysophospholipid signaling in a well-defined model of pathologic aortic injury and observed Angiotensin II (Ang II) increases expression of PLPP3 in SMCs through nuclear factor kappa B (NF-κB) signaling Plpp3 global reduction (Plpp3+/-) or SMC-specific deletion (SM22-Δ) protects hyperlipidemic mice from AngII-mediated aneurysm formation. LPP3 expression regulates SMC differentiation state and lowering LPP3 levels promotes a fibroblast-like phenotype. Decreased inactivation of bioactive lysophosphatidic acid (LPA) in settings of LPP3 deficiency may underlie these phenotypes because deletion of LPA receptor 4 in mice promotes early aortic dilation and rupture in response to AngII. LPP3 expression and LPA signaling influence SMC and vessel wall responses that are important for aortic dissection and aneurysm formation. These findings could have important implications for therapeutics targeting LPA metabolism and signaling in ongoing clinical trials.
Collapse
Affiliation(s)
- Patrick M Van Hoose
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
| | - Liping Yang
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
| | - Maria Kraemer
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
| | - Margo Ubele
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
| | - Andrew J Morris
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
- Lexington Veterans Affair Medical Center, Lexington, KY, USA
| | - Susan S Smyth
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA.
- Lexington Veterans Affair Medical Center, Lexington, KY, USA.
| |
Collapse
|
24
|
Rombouts KB, van Merrienboer TAR, Ket JCF, Bogunovic N, van der Velden J, Yeung KK. The role of vascular smooth muscle cells in the development of aortic aneurysms and dissections. Eur J Clin Invest 2022; 52:e13697. [PMID: 34698377 PMCID: PMC9285394 DOI: 10.1111/eci.13697] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Aortic aneurysms (AA) are pathological dilations of the aorta, associated with an overall mortality rate up to 90% in case of rupture. In addition to dilation, the aortic layers can separate by a tear within the layers, defined as aortic dissections (AD). Vascular smooth muscle cells (vSMC) are the predominant cell type within the aortic wall and dysregulation of vSMC functions contributes to AA and AD development and progression. However, since the exact underlying mechanism is poorly understood, finding potential therapeutic targets for AA and AD is challenging and surgery remains the only treatment option. METHODS In this review, we summarize current knowledge about vSMC functions within the aortic wall and give an overview of how vSMC functions are altered in AA and AD pathogenesis, organized per anatomical location (abdominal or thoracic aorta). RESULTS Important functions of vSMC in healthy or diseased conditions are apoptosis, phenotypic switch, extracellular matrix regeneration and degradation, proliferation and contractility. Stressors within the aortic wall, including inflammatory cell infiltration and (epi)genetic changes, modulate vSMC functions and cause disturbance of processes within vSMC, such as changes in TGF-β signalling and regulatory RNA expression. CONCLUSION This review underscores a central role of vSMC dysfunction in abdominal and thoracic AA and AD development and progression. Further research focused on vSMC dysfunction in the aortic wall is necessary to find potential targets for noninvasive AA and AD treatment options.
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Filiberto AC, Spinosa MD, Elder CT, Su G, Leroy V, Ladd Z, Lu G, Mehaffey JH, Salmon MD, Hawkins RB, Ravichandran KS, Isakson BE, Upchurch GR, Sharma AK. Endothelial pannexin-1 channels modulate macrophage and smooth muscle cell activation in abdominal aortic aneurysm formation. Nat Commun 2022; 13:1521. [PMID: 35315432 PMCID: PMC8938517 DOI: 10.1038/s41467-022-29233-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 03/07/2022] [Indexed: 01/17/2023] Open
Abstract
Pannexin-1 (Panx1) channels have been shown to regulate leukocyte trafficking and tissue inflammation but the mechanism of Panx1 in chronic vascular diseases like abdominal aortic aneurysms (AAA) is unknown. Here we demonstrate that Panx1 on endothelial cells, but not smooth muscle cells, orchestrate a cascade of signaling events to mediate vascular inflammation and remodeling. Mechanistically, Panx1 on endothelial cells acts as a conduit for ATP release that stimulates macrophage activation via P2X7 receptors and mitochondrial DNA release to increase IL-1β and HMGB1 secretion. Secondly, Panx1 signaling regulates smooth muscle cell-dependent intracellular Ca2+ release and vascular remodeling via P2Y2 receptors. Panx1 blockade using probenecid markedly inhibits leukocyte transmigration, aortic inflammation and remodeling to mitigate AAA formation. Panx1 expression is upregulated in human AAAs and retrospective clinical data demonstrated reduced mortality in aortic aneurysm patients treated with Panx1 inhibitors. Collectively, these data identify Panx1 signaling as a contributory mechanism of AAA formation. Pannexin-1 ion channels on endothelial cells regulate vascular inflammation and remodeling to mediate aortic aneurysm formation. Pharmacological blockade of Pannexin-1 channels may offer translational therapeutic mitigation of aneurysmal pathology.
Collapse
|
26
|
Preservation of Smooth Muscle Cell Integrity and Function: A Target for Limiting Abdominal Aortic Aneurysm Expansion? Cells 2022; 11:cells11061043. [PMID: 35326494 PMCID: PMC8947535 DOI: 10.3390/cells11061043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Abdominal aortic aneurysm (AAA) is a silent, progressive disease with significant mortality from rupture. Whilst screening programmes are now able to detect this pathology early in its development, no therapeutic intervention has yet been identified to halt or retard aortic expansion. The inability to obtain aortic tissue from humans at early stages has created a necessity for laboratory models, yet it is essential to create a timeline of events from EARLY to END stage AAA progression. (2) We used a previously validated ex vivo porcine bioreactor model pre-treated with protease enzyme to create “aneurysm” tissue. Mechanical properties, histological changes in the intact vessel wall, and phenotype/function of vascular smooth muscle cells (SMC) cultured from the same vessels were investigated. (3) The principal finding was significant hyperproliferation of SMC from EARLY stage vessels, but without obvious histological or SMC aberrancies. END stage tissue exhibited histological loss of α-smooth muscle actin and elastin; mechanical impairment; and, in SMC, multiple indications of senescence. (4) Aortic SMC may offer a therapeutic target for intervention, although detailed studies incorporating intervening time points between EARLY and END stage are required. Such investigations may reveal mechanisms of SMC dysfunction in AAA development and hence a therapeutic window during which SMC differentiation could be preserved or reinstated.
Collapse
|
27
|
Yang H, DeRoo E, Zhou T, Liu B. Deciphering Cell-Cell Communication in Abdominal Aortic Aneurysm From Single-Cell RNA Transcriptomic Data. Front Cardiovasc Med 2022; 9:831789. [PMID: 35187133 PMCID: PMC8854649 DOI: 10.3389/fcvm.2022.831789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Cell-cell communication coordinates cellular differentiation, tissue homeostasis, and immune responses in states of health and disease. In abdominal aortic aneurysm (AAA), a relatively common and potentially life-threatening vascular disease, intercellular communications between multiple cell types are not fully understood. In this study, we analyzed published single-cell RNA sequencing (scRNA-seq) datasets generated from the murine CaCl2 model, perivascular elastase model, Angiotensin II model, and human AAA using bioinformatic approaches. We inferred the intercellular communication network in each experimental AAA model and human AAA and predicted commonly altered signaling pathways, paying particular attention to thrombospondin (THBS) signaling between different cell populations. Together, our analysis inferred intercellular signaling in AAA based on single-cell transcriptomics. This work provides important insight into cell-cell communications in AAA and has laid the groundwork for future experimental investigations that can elucidate the cell signaling pathways driving AAA.
Collapse
Affiliation(s)
- Huan Yang
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Elise DeRoo
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ting Zhou
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- *Correspondence: Ting Zhou
| | - Bo Liu
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Department of Cellular and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Bo Liu
| |
Collapse
|
28
|
Leveraging cell-type-specific regulatory networks to interpret genetic variants in abdominal aortic aneurysm. Proc Natl Acad Sci U S A 2022; 119:2115601119. [PMID: 34930827 PMCID: PMC8740683 DOI: 10.1073/pnas.2115601119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 12/17/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a common and severe disease with major genetic risk factors. In this study we generated enhancer-promoter contact data to identify regulatory elements in AAA-relevant cell types and identified changes in their predicted chromatin accessibility between AAA patients and controls. We integrated this information with disease-associated variants in regulatory elements and gene bodies to further understand the etiology and pathogenetic mechanisms of AAA. Our study combined whole-genome sequencing data with gene regulatory relations in disease-relevant cell types to reveal the important roles of the interleukin 6 pathway and ERG and KLF regulation in AAA pathogenesis. Abdominal aortic aneurysm (AAA) is a common degenerative cardiovascular disease whose pathobiology is not clearly understood. The cellular heterogeneity and cell-type-specific gene regulation of vascular cells in human AAA have not been well-characterized. Here, we performed analysis of whole-genome sequencing data in AAA patients versus controls with the aim of detecting disease-associated variants that may affect gene regulation in human aortic smooth muscle cells (AoSMC) and human aortic endothelial cells (HAEC), two cell types of high relevance to AAA disease. To support this analysis, we generated H3K27ac HiChIP data for these cell types and inferred cell-type-specific gene regulatory networks. We observed that AAA-associated variants were most enriched in regulatory regions in AoSMC, compared with HAEC and CD4+ cells. The cell-type-specific regulation defined by this HiChIP data supported the importance of ERG and the KLF family of transcription factors in AAA disease. The analysis of regulatory elements that contain noncoding variants and also are differentially open between AAA patients and controls revealed the significance of the interleukin-6-mediated signaling pathway. This finding was further validated by including information from the deleteriousness effect of nonsynonymous single-nucleotide variants in AAA patients and additional control data from the Medical Genome Reference Bank dataset. These results shed important insights into AAA pathogenesis and provide a model for cell-type-specific analysis of disease-associated variants.
Collapse
|
29
|
Hawkins RB, Salmon M, Su G, Lu G, Leroy V, Bontha SV, Mas VR, Jr GRU, Ailawadi G, Sharma AK. Mesenchymal Stem Cells Alter MicroRNA Expression and Attenuate Thoracic Aortic Aneurysm Formation. J Surg Res 2021; 268:221-231. [PMID: 34371281 PMCID: PMC11044812 DOI: 10.1016/j.jss.2021.06.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/13/2021] [Accepted: 06/11/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND Thoracic aortic aneurysms (TAA) are a progressive disease characterized by inflammation, smooth muscle cell activation and matrix degradation. We hypothesized that mesenchymal stem cells (MSCs) can immunomodulate vascular inflammation and remodeling via altered microRNA (miRNAs) expression profile to attenuate TAA formation. MATERIALS AND METHODS C57BL/6 mice underwent topical elastase application to form descending TAAs. Mice were also treated with MSCs on days 1 and 5 and aortas were analyzed on day 14 for aortic diameter. Cytokine array was performed in aortic tissue and total RNA was tagged and hybridized for miRNAs microarray analysis. Immunohistochemistry was performed for elastin degradation and leukocyte infiltration. RESULTS Treatment with MSCs significantly attenuated aortic diameter and TAA formation compared to untreated mice. MSC administration also attenuated T-cell, neutrophil and macrophage infiltration and prevented elastic degradation to mitigate vascular remodeling. MSC treatment also attenuated aortic inflammation by decreasing proinflammatory cytokines (CXCL13, IL-27, CXCL12 and RANTES) and upregulating anti-inflammatory interleukin-10 expression in aortic tissue of elastase-treated mice. TAA formation demonstrated activation of specific miRNAs that are associated with aortic inflammation and vascular remodeling. Our results also demonstrated that MSCs modulate a different set of miRNAs that are associated with decrease leukocyte infiltration and vascular inflammation to attenuate the aortic diameter and TAA formation. CONCLUSIONS These results indicate that MSCs immunomodulate specific miRNAs that are associated with modulating hallmarks of aortic inflammation and vascular remodeling of aortic aneurysms. Targeted therapies designed using MSCs and miRNAs have the potential to regulate the growth and development of TAAs.
Collapse
Affiliation(s)
- Robert B Hawkins
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Morgan Salmon
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Gang Su
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Guanyi Lu
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Victoria Leroy
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Sai Vineela Bontha
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Valeria R Mas
- Department of Surgery, University of Maryland, Baltimore, Maryland
| | | | - Gorav Ailawadi
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Ashish K Sharma
- Department of Surgery, University of Florida, Gainesville, Florida.
| |
Collapse
|
30
|
Gandhi R, Cawthorne C, Craggs LJL, Wright JD, Domarkas J, He P, Koch-Paszkowski J, Shires M, Scarsbrook AF, Archibald SJ, Tsoumpas C, Bailey MA. Cell proliferation detected using [ 18F]FLT PET/CT as an early marker of abdominal aortic aneurysm. J Nucl Cardiol 2021; 28:1961-1971. [PMID: 31741324 PMCID: PMC8648642 DOI: 10.1007/s12350-019-01946-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/17/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a focal aortic dilatation progressing towards rupture. Non-invasive AAA-associated cell proliferation biomarkers are not yet established. We investigated the feasibility of the cell proliferation radiotracer, fluorine-18-fluorothymidine ([18F]FLT) with positron emission tomography/computed tomography (PET/CT) in a progressive pre-clinical AAA model (angiotensin II, AngII infusion). METHODS AND RESULTS Fourteen-week-old apolipoprotein E-knockout (ApoE-/-) mice received saline or AngII via osmotic mini-pumps for 14 (n = 7 and 5, respectively) or 28 (n = 3 and 4, respectively) days and underwent 90-minute dynamic [18F]FLT PET/CT. Organs were harvested from independent cohorts for gamma counting, ultrasound scanning, and western blotting. [18F]FLT uptake was significantly greater in 14- (n = 5) and 28-day (n = 3) AAA than in saline control aortae (n = 5) (P < 0.001), which reduced between days 14 and 28. Whole-organ gamma counting confirmed greater [18F]FLT uptake in 14-day AAA (n = 9) compared to saline-infused aortae (n = 4) (P < 0.05), correlating positively with aortic volume (r = 0.71, P < 0.01). Fourteen-day AAA tissue showed increased expression of thymidine kinase-1, equilibrative nucleoside transporter (ENT)-1, ENT-2, concentrative nucleoside transporter (CNT)-1, and CNT-3 than 28-day AAA and saline control tissues (n = 3 each) (all P < 0.001). CONCLUSIONS [18F]FLT uptake is increased during the active growth phase of the AAA model compared to saline control mice and late-stage AAA.
Collapse
Affiliation(s)
- Richa Gandhi
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, 8.49c Worsley Building, Clarendon Way, Leeds, LS2 9NL, United Kingdom
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds, United Kingdom
| | - Christopher Cawthorne
- Department of Biomedical Science, PET Research Centre, University of Hull, Hull, United Kingdom
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Lucinda J L Craggs
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, 8.49c Worsley Building, Clarendon Way, Leeds, LS2 9NL, United Kingdom
| | - John D Wright
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, 8.49c Worsley Building, Clarendon Way, Leeds, LS2 9NL, United Kingdom
- Experimental & PreClinical Imaging Facility (ePIC), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Juozas Domarkas
- Department of Biomedical Science, PET Research Centre, University of Hull, Hull, United Kingdom
| | - Ping He
- Department of Biomedical Science, PET Research Centre, University of Hull, Hull, United Kingdom
| | - Joanna Koch-Paszkowski
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, 8.49c Worsley Building, Clarendon Way, Leeds, LS2 9NL, United Kingdom
- Experimental & PreClinical Imaging Facility (ePIC), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Michael Shires
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Andrew F Scarsbrook
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Stephen J Archibald
- Department of Biomedical Science, PET Research Centre, University of Hull, Hull, United Kingdom
| | - Charalampos Tsoumpas
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, 8.49c Worsley Building, Clarendon Way, Leeds, LS2 9NL, United Kingdom.
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Invicro, London, United Kingdom.
| | - Marc A Bailey
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, 8.49c Worsley Building, Clarendon Way, Leeds, LS2 9NL, United Kingdom
- The Leeds Vascular Institute, Leeds General Infirmary, Great George Street, Leeds, United Kingdom
| |
Collapse
|
31
|
Dou X, Ma Y, Qin Y, Dong Q, Zhang S, Tian R, Pan M. NEAT1 silencing alleviates pulmonary arterial smooth muscle cell migration and proliferation under hypoxia through regulation of miR‑34a‑5p/KLF4 in vitro. Mol Med Rep 2021; 24:749. [PMID: 34468014 DOI: 10.3892/mmr.2021.12389] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/29/2021] [Indexed: 11/06/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe vascular disease that adversely affects patient health and can be life threatening. The present study aimed to investigate the detailed role of nuclear paraspeckle assembly transcript 1 (NEAT1) in PAH. Using RT‑qPCR, the expression levels of NEAT1, microRNA (miR)‑34a‑5p, and Krüppel‑like factor 4 (KLF4) were detected in both hypoxia‑treated pulmonary arterial smooth muscle cells (PASMCs) and serum from PAH patients. Then, the interactions among miR‑34a‑5p, NEAT1, and KLF4 were evaluated by dual‑luciferase reporter assay. The detailed role of the NEAT1/miR‑34a‑5p/KLF4 axis in PAH pathogenesis was further explored using MTT, Transwell, and western blot assays. The results revealed that NEAT1 targeted miR‑34a‑5p and miR‑34a‑5p targeted KLF4. In hypoxia‑treated PASMCs and serum from PAH patients, high NEAT1 and KLF4 expression levels and low miR‑34a‑5p expression were observed. The proliferation and migration of hypoxia‑treated PASMCs were reduced by transfection with sh‑NEAT1 or miR‑34a‑5p mimics. The suppressive effects of NEAT1 knockdown on the proliferation and migration of hypoxia‑treated PASMCs were reversed by knock down of miR‑34a‑5p expression and increased KLF4 expression. NEAT1 was not only highly expressed in the serum of PAH patients but its silencing also alleviated PAH by regulating miR‑34a‑5p/KLF4 in vitro. The present study highlighted a potential new therapeutic target and diagnostic biomarker for PAH.
Collapse
Affiliation(s)
- Xiuli Dou
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Yuxiao Ma
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Yijie Qin
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Qinglin Dong
- Department of Emergency, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Shouwei Zhang
- Department of Medical, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Rui Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Mingyu Pan
- Department of Cardiology, The People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| |
Collapse
|
32
|
Liu H, Zhang Y, Song W, Sun Y, Jiang Y. Osteopontin N-Terminal Function in an Abdominal Aortic Aneurysm From Apolipoprotein E-Deficient Mice. Front Cell Dev Biol 2021; 9:681790. [PMID: 34458254 PMCID: PMC8397420 DOI: 10.3389/fcell.2021.681790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/20/2021] [Indexed: 12/20/2022] Open
Abstract
The cleavage of osteopontin (OPN) by thrombin results in an N-terminal fragment (OPN-N), which exposes a cryptic integrin-binding motif that promotes the adherence of cells, and plays a proinflammatory role. However, the effect of OPN-N on abdominal aortic aneurysm (AAA) remains unknown. The aim of this study was to investigate the expression of OPN-N in aortic tissue samples obtained from patients, who underwent acute aortic dissection (AD), and normal aorta, effect of OPN-N on angiotensin (Ang) II-induced AAA in mice, and relationship between OPN-N and pyroptosis-related inflammatory factors in vitro. Hematoxylin and eosin staining was conducted to detect histological changes. Next, we detected the expression of the OPN-N protein. Additionally, ApoE−/− mice were divided into four groups: control, control + M5Ab (to block the OPN-N function in mice), Ang II, and Ang II + M5Ab. All mice were euthanized after a 28-day infusion and whole aortas, including thoracic and abdominal aortas, were collected for morphological and histological analysis of the AAA. The OPN-N protein expression was higher in patients with AD than in normal individuals, while histological changes in the aortas of Ang II mice were suppressed in Ang II + M5Ab mice. The expression of OPN-N, NOD-, LRR-, and pyrin domain-containing protein 3, pro-Caspase-1, ASC, Gasdermin-d, interleukin (IL)-18, IL-1β, matrix metalloproteinase (MMP) 2, and MMP9 was lower in the Ang II + M5Ab group than in the Ang II group. The gene expression of monocyte chemoattractant protein-1, IL-6, and tumor necrosis factor-α was suppressed in the aortic tissues of the Ang II + M5Ab group compared with the Ang II group. Moreover, the expression of α-smooth muscle actin was lower in the Ang II group than in the Ang II + M5Ab group. In vitro results showed that the increase in the expression of pyroptosis-related inflammatory factors induced by OPN was mediated through the nuclear factor (NF)-κB pathway. In conclusion, OPN-N promotes AAA by increasing the expression of pyroptosis-related inflammatory factors through the NF-κB pathway, inflammation, and extracellular matrix degradation. These results highlight the potential of OPN-N as a new therapeutic target to prevent AAA expansion.
Collapse
Affiliation(s)
- Hongyang Liu
- Department of Heart Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Zhang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Song
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yancui Sun
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yinong Jiang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
33
|
Liu X, Guo Y, Yang Y, Qi C, Xiong T, Chen Y, Wu G, Zeng C, Wang D. DRD4 (Dopamine D4 Receptor) Mitigate Abdominal Aortic Aneurysm via Decreasing P38 MAPK (mitogen-activated protein kinase)/NOX4 (NADPH Oxidase 4) Axis-Associated Oxidative Stress. Hypertension 2021; 78:294-307. [PMID: 34176291 DOI: 10.1161/hypertensionaha.120.16738] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Xuesong Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China (X.L., C.Q., T.X.)
| | - Yansong Guo
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fuzhou, China (Y.G.)
| | - Yuxue Yang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), China (Y.Y., D.W.)
| | - Chunlei Qi
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China (X.L., C.Q., T.X.)
| | - Ting Xiong
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China (X.L., C.Q., T.X.)
| | - Yue Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China (Y.C., G.W., C.Z.)
| | - Gengze Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China (Y.C., G.W., C.Z.)
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China (Y.C., G.W., C.Z.)
| | - Daxin Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), China (Y.Y., D.W.)
| |
Collapse
|
34
|
Xiong JM, Liu H, Chen J, Zou QQ, Wang YYJ, Bi GS. Curcumin nicotinate suppresses abdominal aortic aneurysm pyroptosis via lncRNA PVT1/miR-26a/KLF4 axis through regulating the PI3K/AKT signaling pathway. Toxicol Res (Camb) 2021; 10:651-661. [PMID: 34141179 DOI: 10.1093/toxres/tfab041] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/19/2021] [Accepted: 04/08/2021] [Indexed: 11/15/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic dilated disease of the aorta that is characterized by chronic inflammation. Curcumin (Cur) is previously showed to attenuate AAA by inhibiting inflammatory response in ApoE -/- mice. Since Cur has the limitations of aqueous solubility and instability. Here, we focus on the role of curcumin nicotinate (CurTn), a Cur derivative is derived from Cur and nicotinate. An in vitro model of AAA was established by treating vascular smooth muscle cells (VSMCs) with II (Ang-II). Gene and protein expressions were examined by quantitative real-time PCR (qPCR) or western blotting. Cell migration and pyroptosis were determined by transwell assay and flow cytometry. The interaction between plasmacytoma variant translocation 1 (PVT1), miR-26a and krüppel-like factor 4 (KLF4) was predicted by online prediction tool and confirmed by luciferase reporter assay. CurTn reduced Ang-II-induced AAA-associated proteins, inflammatory cytokine expressions, and attenuated pyroptosis in VSMCs. PVT1 overexpression suppressed the inhibitory effect of CurTn on AngII-induced pyroptosis and inflammatory in VSMCs by sponging miR-26a. miR-26a directly targeted KLF4 and suppressed its expression, which eventually led to the deactivation of the PI3K/AKT signaling pathway. Besides, the regulatory effect of CurTn on pyroptosis of VSMCs induced by Ang-II was reversed through the PVT1/miR-26a/KLF4 pathway. In short, CurTn suppressed VSMCs pyroptosis and inflammation though mediation PVT1/miR-26a/KLF4 axis by regulating the PI3K/AKT signaling pathway, CurTn might consider as a potential therapeutic target in the treatment of AAA.
Collapse
Affiliation(s)
- Jian-Ming Xiong
- Department of Vascular Surgery, Yiyang Central Hospital, Yiyang 413000, Hunan Province, P.R. China
| | - Hui Liu
- Department of Vascular Surgery, Yiyang Central Hospital, Yiyang 413000, Hunan Province, P.R. China
| | - Jie Chen
- Department of Vascular Surgery, The Second Affiliated Hospital, University of South China, Hengyang 421000, Hunan Province, P.R. China
| | - Qing-Qing Zou
- Department of Vascular Surgery, The Second Affiliated Hospital, University of South China, Hengyang 421000, Hunan Province, P.R. China
| | - Yang-Yi-Jing Wang
- Department of Vascular Surgery, The Second Affiliated Hospital, University of South China, Hengyang 421000, Hunan Province, P.R. China
| | - Guo-Shan Bi
- Department of Vascular Surgery, The Second Affiliated Hospital, University of South China, Hengyang 421000, Hunan Province, P.R. China
| |
Collapse
|
35
|
Lu W, Zhou Y, Zeng S, Zhong L, Zhou S, Song H, Ding R, Zhong G, Li Q, Hu Y, Wen Z, Liao Q, Wang Y, Lyu L, Zhong Y, Hu G, Liao Y, Xie D, Xie J. Loss of FoxO3a prevents aortic aneurysm formation through maintenance of VSMC homeostasis. Cell Death Dis 2021; 12:378. [PMID: 33828087 PMCID: PMC8027644 DOI: 10.1038/s41419-021-03659-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 11/09/2022]
Abstract
Vascular smooth muscle cell (VSMC) phenotypic switching plays a critical role in the formation of abdominal aortic aneurysms (AAAs). FoxO3a is a key suppressor of VSMC homeostasis. We found that in human and animal AAA tissues, FoxO3a was upregulated, SM22α and α-smooth muscle actin (α-SMA) proteins were downregulated and synthetic phenotypic markers were upregulated, indicating that VSMC phenotypic switching occurred in these diseased tissues. In addition, in cultured VSMCs, significant enhancement of FoxO3a expression was found during angiotensin II (Ang II)-induced VSMC phenotypic switching. In vivo, FoxO3a overexpression in C57BL/6J mice treated with Ang II increased the formation of AAAs, whereas FoxO3a knockdown exerted an inhibitory effect on AAA formation in ApoE−/− mice infused with Ang II. Mechanistically, FoxO3a overexpression significantly inhibited the expression of differentiated smooth muscle cell (SMC) markers, activated autophagy, the essential repressor of VSMC homeostasis, and promoted AAA formation. Our study revealed that FoxO3a promotes VSMC phenotypic switching to accelerate AAA formation through the P62/LC3BII autophagy signaling pathway and that therapeutic approaches that decrease FoxO3a expression may prevent AAA formation.
Collapse
Affiliation(s)
- Weiling Lu
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China.,Department of Cardiology, Ganzhou Municipal Hospital, 49th, Grand Highway, 341000, Ganzhou, China
| | - Yu Zhou
- Division of Vascular Surgery, National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangdong Engineering Laboratory of Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shan Zeng
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Lintao Zhong
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), 519000, Zhuhai, China
| | - Shiju Zhou
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Haoyu Song
- Wards of Cadres, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), 519000, Zhuhai, China
| | - Rongming Ding
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Gaojun Zhong
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Qingrui Li
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Yuhua Hu
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Zhongyu Wen
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Qin Liao
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Yalan Wang
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Lianglliang Lyu
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Yiming Zhong
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Gonghua Hu
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515, Guangzhou, China.
| | - Dongming Xie
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China. .,Jiangxi Branch Center of National Geriatric Disease Clinical Medical Research Center, Gannan Medical University, University Town, 341000, Ganzhou Development District, Jiangxi Province, China.
| | - Jiahe Xie
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, 341000, Ganzhou, China. .,Jiangxi Branch Center of National Geriatric Disease Clinical Medical Research Center, Gannan Medical University, University Town, 341000, Ganzhou Development District, Jiangxi Province, China.
| |
Collapse
|
36
|
Bell M, Gandhi R, Shawer H, Tsoumpas C, Bailey MA. Imaging Biological Pathways in Abdominal Aortic Aneurysms Using Positron Emission Tomography. Arterioscler Thromb Vasc Biol 2021; 41:1596-1606. [PMID: 33761759 DOI: 10.1161/atvbaha.120.315812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Michael Bell
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Richa Gandhi
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Heba Shawer
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Charalampos Tsoumpas
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Marc A Bailey
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| |
Collapse
|
37
|
SM22 α Loss Contributes to Apoptosis of Vascular Smooth Muscle Cells via Macrophage-Derived circRasGEF1B. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5564884. [PMID: 33859778 PMCID: PMC8026322 DOI: 10.1155/2021/5564884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/24/2021] [Accepted: 03/02/2021] [Indexed: 11/18/2022]
Abstract
Vascular smooth muscle cell (VSMC) apoptosis is a major defining feature of abdominal aortic aneurysm (AAA) and mainly caused by inflammatory cell infiltration. Smooth muscle (SM) 22α prevents AAA formation through suppressing NF-κB activation. However, the role of SM22α in VSMC apoptosis is controversial. Here, we identified that SM22α loss contributed to apoptosis of VSMCs via activation of macrophages. Firstly, deficiency of SM22α enhanced the interaction of VSMCs with macrophages. Macrophages were retained and activated by Sm22α−/− VSMCs via upregulating VCAM-1 expression. The ratio of apoptosis was increased by 1.62-fold in VSMCs treated with the conditional media (CM) from activated RAW264.7 cells, compared to that of the control CM (P < 0.01), and apoptosis of Sm22α−/− VSMCs was higher than that of WT VSMCs (P < 0.001). Next, circRasGEF1B from activated macrophages was delivered into VSMCs promoting ZFP36 expression via stabilization of ZFP36 mRNA. Importantly, circRasGEF1B, as a scaffold, guided ZFP36 to preferentially bind to and decay Bcl-2 mRNA in a sequence-specific manner and triggered apoptosis of VSMCs, especially in Sm22α−/− VSMCs. These findings reveal a novel mechanism by which the circRasGEF1B-ZFP36 axis mediates macrophage-induced VSMC apoptosis via decay of Bcl-2 mRNA, whereas Sm22α−/− VSMCs have a higher sensitivity to apoptosis.
Collapse
|
38
|
Si X, Chen Q, Zhang J, Zhou W, Chen L, Chen J, Deng N, Li W, Liu D, Wang L, Shi L, Sun W, Song H, Zhong L. MicroRNA-23b prevents aortic aneurysm formation by inhibiting smooth muscle cell phenotypic switching via FoxO4 suppression. Life Sci 2021; 288:119092. [PMID: 33737086 DOI: 10.1016/j.lfs.2021.119092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/10/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
AIMS Phenotypic switching of vascular smooth muscle cells (VSMCs) is essential for the formation of abdominal aortic aneurysms (AAAs). MicroRNA-23b (miR-23b) has recently been shown to play a vital role in maintaining the VSMC contractile phenotype; however, little is known about the role of miR-23b in the formation of AAAs. Here, we investigated whether miR-23b prevents AAA formation by inhibiting VSMC phenotypic switching. MATERIALS AND METHODS We administered angiotensin II (Ang II, 1000 ng/kg/min) or vehicle to 10-12-week-old male apolipoprotein E knockout (ApoE-/-) or C57BL/6J mice via subcutaneous osmotic minipumps for 4 weeks. KEY FINDINGS The expression of miR-23b was significantly reduced in the aorta during the early onset of AAA in angiotensin II-treated ApoE-/- mice and in human AAA samples. In vitro experiments showed that the suppression of SMC contractile marker gene expression induced by Ang II was accelerated by miR-23b inhibitors but inhibited by mimics. In vivo studies revealed that miR-23b deficiency in Ang II-treated C57BL/6J mice aggravated the formation of AAAs in these mice compared with control mice; the opposite results were observed in miR-23b-overexpressing mice. Mechanistically, miR-23b knockdown significantly increased the expression of the transcription factor forkhead box O4 (FoxO4) during VSMC phenotypic switching induced by Ang II. In addition, a luciferase reporter assay showed that FoxO4 is a target of miR-23b in VSMCs. SIGNIFICANCE Our study revealed a pivotal role for miR-23b in protecting against aortic aneurysm formation by maintaining the VSMC contractile phenotype.
Collapse
Affiliation(s)
- Xiaoyun Si
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China; Geriatrics Department, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China
| | - Qixian Chen
- Department of Pulmonary and Critical Care Medicines, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China
| | - Jiechang Zhang
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China
| | - Wei Zhou
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Lijun Chen
- Department of Hematology and Rheumatology, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China
| | - Jingjing Chen
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Na Deng
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Wei Li
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Danan Liu
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Long Wang
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Linyan Shi
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Weihong Sun
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Haoyu Song
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, Ganzhou 341000, China.
| | - Lintao Zhong
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China.
| |
Collapse
|
39
|
Cooper HA, Cicalese S, Preston KJ, Kawai T, Okuno K, Choi ET, Kasahara S, Uchida HA, Otaka N, Scalia R, Rizzo V, Eguchi S. Targeting mitochondrial fission as a potential therapeutic for abdominal aortic aneurysm. Cardiovasc Res 2021; 117:971-982. [PMID: 32384150 PMCID: PMC7898955 DOI: 10.1093/cvr/cvaa133] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/15/2020] [Accepted: 04/30/2020] [Indexed: 11/12/2022] Open
Abstract
AIMS Angiotensin II (AngII) is a potential contributor to the development of abdominal aortic aneurysm (AAA). In aortic vascular smooth muscle cells (VSMCs), exposure to AngII induces mitochondrial fission via dynamin-related protein 1 (Drp1). However, pathophysiological relevance of mitochondrial morphology in AngII-associated AAA remains unexplored. Here, we tested the hypothesis that mitochondrial fission is involved in the development of AAA. METHODS AND RESULTS Immunohistochemistry was performed on human AAA samples and revealed enhanced expression of Drp1. In C57BL6 mice treated with AngII plus β-aminopropionitrile, AAA tissue also showed an increase in Drp1 expression. A mitochondrial fission inhibitor, mdivi1, attenuated AAA size, associated aortic pathology, Drp1 protein induction, and mitochondrial fission but not hypertension in these mice. Moreover, western-blot analysis showed that induction of matrix metalloproteinase-2, which precedes the development of AAA, was blocked by mdivi1. Mdivi1 also reduced the development of AAA in apolipoprotein E-deficient mice infused with AngII. As with mdivi1, Drp1+/- mice treated with AngII plus β-aminopropionitrile showed a decrease in AAA compared to control Drp1+/+ mice. In abdominal aortic VSMCs, AngII induced phosphorylation of Drp1 and mitochondrial fission, the latter of which was attenuated with Drp1 silencing as well as mdivi1. AngII also induced vascular cell adhesion molecule-1 expression and enhanced leucocyte adhesion and mitochondrial oxygen consumption in smooth muscle cells, which were attenuated with mdivi1. CONCLUSION These data indicate that Drp1 and mitochondrial fission play salient roles in AAA development, which likely involves mitochondrial dysfunction and inflammatory activation of VSMCs.
Collapse
MESH Headings
- Aminopropionitrile
- Angiotensin II
- Animals
- Anti-Inflammatory Agents/pharmacology
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Case-Control Studies
- Cell Adhesion/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dynamins/genetics
- Dynamins/metabolism
- Humans
- Leukocytes/drug effects
- Leukocytes/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/genetics
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Mitochondrial Dynamics/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oxygen Consumption/drug effects
- Phosphorylation
- Quinazolinones/pharmacology
- Mice
Collapse
Affiliation(s)
- Hannah A Cooper
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Stephanie Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Kyle J Preston
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Keisuke Okuno
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Eric T Choi
- Department of Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Shingo Kasahara
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Haruhito A Uchida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nozomu Otaka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
40
|
He J, Li N, Fan Y, Zhao X, Liu C, Hu X. Metformin Inhibits Abdominal Aortic Aneurysm Formation through the Activation of the AMPK/mTOR Signaling Pathway. J Vasc Res 2021; 58:148-158. [PMID: 33601368 DOI: 10.1159/000513465] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/26/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Epidemiological evidence suggests that the antidiabetic drug metformin (MET) can also inhibit abdominal aortic aneurysm (AAA) formation. However, the underlying protective mechanism remains unknown. It has been reported that phosphorylated AMP-activated protein kinase (AMPK) levels are significantly lower in AAA tissues than control aortic tissues. AMPK activation can inhibit the downstream signaling molecule called mechanistic target of rapamycin (mTOR), which has also been reported be upregulated in thoracic aneurysms. Thus, blocking mTOR signaling could attenuate AAA progression. MET is a known agonist of AMPK. Therefore, in this study, we investigated if MET could inhibit formation of AAA by activating the AMPK/mTOR signaling pathway. MATERIALS AND METHODS The AAA animal model was induced by intraluminal porcine pancreatic elastase (PPE) perfusion in male Sprague Dawley rats. The rats were treated with MET or compound C (C.C), which is an AMPK inhibitor. AAA formation was monitored by serial ultrasound. Aortas were collected 4 weeks after surgery and subjected to immunohistochemistry, Western blot, and transmission electron microscopy analyses. RESULTS MET treatment dramatically inhibited the formation of AAA 4 weeks after PPE perfusion. MET reduced the aortic diameter, downregulated both macrophage infiltration and matrix metalloproteinase expression, decreased neovascularization, and preserved the contractile phenotype of the aortic vascular smooth muscle cells. Furthermore, we detected an increase in autophagy after MET treatment. All of these effects were reversed by the AMPK inhibitor C.C. CONCLUSION This study demonstrated that MET activates AMPK and suppresses AAA formation. Our study provides a novel mechanism for MET and suggests that MET could be potentially used as a therapeutic candidate for preventing AAA.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/enzymology
- Aorta, Abdominal/ultrastructure
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Dilatation, Pathologic
- Disease Models, Animal
- Enzyme Activation
- Macrophages/drug effects
- Macrophages/metabolism
- Male
- Metformin/pharmacology
- Neovascularization, Pathologic
- Pancreatic Elastase
- Phosphorylation
- Rats, Sprague-Dawley
- Signal Transduction
- TOR Serine-Threonine Kinases/metabolism
- Vascular Remodeling/drug effects
- Rats
Collapse
Affiliation(s)
- Jiaan He
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Nan Li
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yichuan Fan
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xingzhi Zhao
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chengwei Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Xinhua Hu
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang, China,
| |
Collapse
|
41
|
Time-Dependent Pathological Changes in Hypoperfusion-Induced Abdominal Aortic Aneurysm. BIOLOGY 2021; 10:biology10020149. [PMID: 33672844 PMCID: PMC7917844 DOI: 10.3390/biology10020149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 11/19/2022]
Abstract
Simple Summary Abdominal aortic aneurysm (AAA) is a vascular disease that involves gradual dilation of the abdominal aorta and has a high mortality due to rupture. Hypoperfusion due to the obstruction of vasa vasorum, which is a blood supply system in the aortic wall, may be an important factor involved in AAA pathophysiology. A time-dependent analysis is important to understand the pathological cascade following hypoperfusion in the aortic wall. In our study, time-dependent analysis using a hypoperfusion-induced animal model showed that the dynamics of many AAA-related factors might be associated with the increased hypoxia-inducible factor-1α level. Hypoperfusion due to stenosis of the vasa vasorum might be a new drug target for AAA therapeutics. Abstract Hypoperfusion due to vasa vasorum stenosis can cause wall hypoxia and abdominal aortic aneurysm (AAA) development. Even though hypoperfusion is an important contributor toward pathological changes in AAA, the correlation between hypoperfusion and AAA is not fully understood. In this study, a time-dependent semi-quantitative pathological analysis of hypoperfusion-induced aortic wall changes was performed to understand the mechanisms underlying the gradual degradation of the aortic wall leading to AAA formation. AAA-related factors evaluated in this study were grouped according to the timing of dynamic change, and five groups were formed as follows: first group: angiotensin II type 1 receptor, endothelin-1 (ET-1), and malondialdehyde (MDA); second group: matrix metalloproteinase (MMP)-2, -9, -12, M1 macrophages (Mac387+ cells), and monocyte chemotactic protein-1; third group: synthetic smooth muscle cells (SMCs); fourth group: neutrophil elastase, contractile SMCs, and angiotensinogen; and the fifth group: M2 macrophages (CD163+ cells). Hypoxia-inducible factor-1α, ET-1, MDA, and MMP-9 were colocalized with alpha-smooth muscle actin cells in 3 h, suggesting that hypoperfusion-induced hypoxia directly affects the activities of contractile SMCs in the initial stage of AAA. Time-dependent pathological analysis clarified the cascade of AAA-related factors. These findings provide clues for understanding complicated multistage pathologies in AAA.
Collapse
|
42
|
Bernal S, Lopez-Sanz L, Jimenez-Castilla L, Prieto I, Melgar A, La Manna S, Martin-Ventura JL, Blanco-Colio LM, Egido J, Gomez-Guerrero C. Protective effect of suppressor of cytokine signalling 1-based therapy in experimental abdominal aortic aneurysm. Br J Pharmacol 2020; 178:564-581. [PMID: 33227156 DOI: 10.1111/bph.15330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 09/20/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Abdominal aortic aneurysm (AAA) is a multifactorial disease characterized by chronic inflammation, oxidative stress and proteolytic activity in the aortic wall. Targeting JAK/signal transducer and activator of transcription (JAK/STAT) pathway is a promising strategy for chronic inflammatory diseases. We investigated the vasculo-protective role of suppressor of cytokine signalling-1 (SOCS1), the negative JAK/STAT regulator, in experimental AAA. EXPERIMENTAL APPROACH A synthetic, cell permeable peptide (S1) mimic of SOCS1 kinase inhibitory domain to suppress STAT activation was evaluated in the well-established mouse model of elastase-induced AAA by monitoring changes in aortic diameter, cellular composition and gene expression in abdominal aorta. S1 function was further evaluated in cultured vascular smooth muscle cells (VSMC) and macrophages exposed to elastase or elastin-derived peptides. KEY RESULTS S1 peptide prevented AAA development, evidenced by reduced incidence of AAA, aortic dilation and elastin degradation, partial restoration of medial VSMC and decreased inflammatory cells and oxidative stress in AAA tissue. Mechanistically, S1 suppressed STAT1/3 activation in aorta, down-regulated cytokines, metalloproteinases and altered the expression of cell differentiation markers by favouring anti-inflammatory M2 macrophage and contractile VSMC phenotypes. In vitro, S1 suppressed the expression of inflammatory and oxidative genes, reduced cell migration and reversed the phenotypic switch of macrophages and VSMC. By contrast, SOCS1 silencing promoted inflammatory response. CONCLUSION AND IMPLICATIONS This preclinical study demonstrates the therapeutic potential of SOCS1-derived peptide to halt AAA progression by suppressing JAK/STAT-mediated inflammation and aortic dilation. S1 peptide may therefore be a valuable option for the treatment of AAA.
Collapse
Affiliation(s)
- Susana Bernal
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Laura Lopez-Sanz
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Luna Jimenez-Castilla
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Ignacio Prieto
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Ana Melgar
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain
| | - Sara La Manna
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain
| | - Jose Luis Martin-Ventura
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Luis Miguel Blanco-Colio
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Jesus Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Carmen Gomez-Guerrero
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz (IIS-FJD), Autonoma University of Madrid (UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| |
Collapse
|
43
|
Wang S, Tian X, Liu D, Zhang X, Yan C, Han Y. TRPV5 attenuates abdominal aortic aneurysm in mice by regulating KLF4-dependent phenotype switch of aortic vascular smooth muscle cells. Arch Biochem Biophys 2020; 698:108724. [PMID: 33309615 DOI: 10.1016/j.abb.2020.108724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a fatal vascular disease with insidious symptoms. However, the mechanism behind its development remains unclear. The transient receptor potential vanilloid (TRPV) family has crucial protective effects against cardiovascular diseases, but the role of TRPV5 in AAA has yet to be reported. In this study, ApoE-/- mice were intraperitoneally injected with AAV-GFP or AAV-TRPV5. After 30 days, mice were further administered with angiotensin II (Ang II, 1.44 mg/kg/day) by using osmotic pumps to induce the AAA model or Saline for 28 days, (i.e., Saline + AAV-GFP, Saline + AAV-TRPV5, Ang II + AAV-GFP and Ang II + AAV-TRPV5 groups were established). Compared with the control group, the incidence of AAA and the maximal diameter of the abdominal aorta markedly decreased in Ang II + AAV-TRPV5, which was detected by vascular ultrasound at 28 day. Meanwhile, less collagen and elastin degradation were observed in the Ang II + AAV-TRPV5 group by using Masson and Elastin stains. Moreover, more α-SMA and less MMP2 was observed in the abdominal aortas collected at 28 day by immunohistochemistry. In vitro, primary mouse vascular smooth muscle cells (VSMCs) were treated with Ang II (1 μM) to induce phenotype switch. Sh-TRPV5 and AdTRPV5 were used to transfect VSMCs. PCR and Western blotting were used to access the expression of contractile marker, including α-SMA and SM-22α. The results showed that the mRNA and protein level of α-SMA and SM-22α were decreased under the stimulation of Ang II, but could be attenuated by TRPV5 overexpression. The cell scratch assay demonstrated that the migration ability of VSMCs was increased in Ang II treated group and could be ameliorated by TRPV5 overexpression. Above all, VSMCs transformed from the contractile into secretory phenotype under Ang II stimuli, but could be rescued by TRPV5 overexpression. Furthermore, TRPV5 overexpression suppressed the increased expression of KLF4 induced by Ang II treatment in VSMCs. The data demonstrated that TRPV5 could inhibit AAA formation and play a critical role in the VSMC phenotype switch by downregulating KLF4, suggesting TRPV5 as a new strategy for treating AAA.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China; Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaoxiang Tian
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaolin Zhang
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China; Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
44
|
Lu S, Jolly AJ, Strand KA, Dubner AM, Mutryn MF, Moulton KS, Nemenoff RA, Majesky MW, Weiser-Evans MC. Smooth muscle-derived progenitor cell myofibroblast differentiation through KLF4 downregulation promotes arterial remodeling and fibrosis. JCI Insight 2020; 5:139445. [PMID: 33119549 PMCID: PMC7714399 DOI: 10.1172/jci.insight.139445] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Resident vascular adventitial SCA1+ progenitor (AdvSca1) cells are essential in vascular development and injury. However, the heterogeneity of AdvSca1 cells presents a unique challenge in understanding signaling pathways orchestrating their behavior in homeostasis and injury responses. Using smooth muscle cell (SMC) lineage-tracing models, we identified a subpopulation of AdvSca1 cells (AdvSca1-SM) originating from mature SMCs that undergo reprogramming in situ and exhibit a multipotent phenotype. Here we employed lineage tracing and RNA-sequencing to define the signaling pathways regulating SMC-to-AdvSca1-SM cell reprogramming and AdvSca1-SM progenitor cell phenotype. Unbiased hierarchical clustering revealed that genes related to hedgehog/WNT/beta-catenin signaling were significantly enriched in AdvSca1-SM cells, emphasizing the importance of this signaling axis in the reprogramming event. Leveraging AdvSca1-SM–specific expression of GLI-Kruppel family member GLI1 (Gli1), we generated Gli1-CreERT2-ROSA26-YFP reporter mice to selectively track AdvSca1-SM cells. We demonstrated that physiologically relevant vascular injury or AdvSca1-SM cell–specific Kruppel-like factor 4 (Klf4) depletion facilitated the proliferation and differentiation of AdvSca1-SM cells to a profibrotic myofibroblast phenotype rather than macrophages. Surprisingly, AdvSca1-SM cells selectively contributed to adventitial remodeling and fibrosis but little to neointima formation. Together, these findings strongly support therapeutics aimed at preserving the AdvSca1-SM cell phenotype as a viable antifibrotic approach. Smooth muscle cell–derived resident vascular adventitial progenitor cells adopt a myofibroblast phenotype in response to vascular injury and play a dominant role in vascular fibrosis.
Collapse
Affiliation(s)
- Sizhao Lu
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Austin J Jolly
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Keith A Strand
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Allison M Dubner
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Marie F Mutryn
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | | | - Raphael A Nemenoff
- Division of Renal Diseases and Hypertension, Department of Medicine, and.,Consortium for Fibrosis Research and Translation, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark W Majesky
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Pediatrics and Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Mary Cm Weiser-Evans
- Division of Renal Diseases and Hypertension, Department of Medicine, and.,Consortium for Fibrosis Research and Translation, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Cardio Vascular Pulmonary Research Lab, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
45
|
Abstract
Macrophages have a key functional role in the pathogenesis of various cardiovascular diseases, such as atherosclerosis and aortic aneurysms. Their accumulation within the vessel wall leads to sustained local inflammatory responses characterized by secretion of chemokines, cytokines, and matrix protein degrading enzymes. Here, we summarize some recent findings on macrophage contribution to cardiovascular disease. We focus on the origin, survival/death, and phenotypic switching of macrophages within vessel walls.
Collapse
Affiliation(s)
- Mitri K Khoury
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Wisconsin, Madison
| | - Huan Yang
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Wisconsin, Madison
| | - Bo Liu
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Wisconsin, Madison
| |
Collapse
|
46
|
Increased Serum KLF4 in Severe Atheromatosis and Extensive Aneurysmal Disease. Ann Vasc Surg 2020; 68:338-343. [DOI: 10.1016/j.avsg.2020.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 11/18/2022]
|
47
|
Pedroza AJ, Tashima Y, Shad R, Cheng P, Wirka R, Churovich S, Nakamura K, Yokoyama N, Cui JZ, Iosef C, Hiesinger W, Quertermous T, Fischbein MP. Single-Cell Transcriptomic Profiling of Vascular Smooth Muscle Cell Phenotype Modulation in Marfan Syndrome Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2020; 40:2195-2211. [PMID: 32698686 PMCID: PMC7484233 DOI: 10.1161/atvbaha.120.314670] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To delineate temporal and spatial dynamics of vascular smooth muscle cell (SMC) transcriptomic changes during aortic aneurysm development in Marfan syndrome (MFS). Approach and Results: We performed single-cell RNA sequencing to study aortic root/ascending aneurysm tissue from Fbn1C1041G/+ (MFS) mice and healthy controls, identifying all aortic cell types. A distinct cluster of transcriptomically modulated SMCs (modSMCs) was identified in adult Fbn1C1041G/+ mouse aortic aneurysm tissue only. Comparison with atherosclerotic aortic data (ApoE-/- mice) revealed similar patterns of SMC modulation but identified an MFS-specific gene signature, including plasminogen activator inhibitor-1 (Serpine1) and Kruppel-like factor 4 (Klf4). We identified 481 differentially expressed genes between modSMC and SMC subsets; functional annotation highlighted extracellular matrix modulation, collagen synthesis, adhesion, and proliferation. Pseudotime trajectory analysis of Fbn1C1041G/+ SMC/modSMC transcriptomes identified genes activated differentially throughout the course of phenotype modulation. While modSMCs were not present in young Fbn1C1041G/+ mouse aortas despite small aortic aneurysm, multiple early modSMCs marker genes were enriched, suggesting activation of phenotype modulation. modSMCs were not found in nondilated adult Fbn1C1041G/+ descending thoracic aortas. Single-cell RNA sequencing from human MFS aortic root aneurysm tissue confirmed analogous SMC modulation in clinical disease. Enhanced expression of TGF-β (transforming growth factor beta)-responsive genes correlated with SMC modulation in mouse and human data sets. CONCLUSIONS Dynamic SMC phenotype modulation promotes extracellular matrix substrate modulation and aortic aneurysm progression in MFS. We characterize the disease-specific signature of modSMCs and provide temporal, transcriptomic context to the current understanding of the role TGF-β plays in MFS aortopathy. Collectively, single-cell RNA sequencing implicates TGF-β signaling and Klf4 overexpression as potential upstream drivers of SMC modulation.
Collapse
Affiliation(s)
- Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Yasushi Tashima
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Rohan Shad
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine. Stanford CA, USA
| | - Robert Wirka
- Division of Cardiovascular Medicine, Stanford University School of Medicine. Stanford CA, USA
| | - Samantha Churovich
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Ken Nakamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Nobu Yokoyama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Jason Z. Cui
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Cristiana Iosef
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine. Stanford CA, USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| |
Collapse
|
48
|
Chen X, Li Y, Xiao J, Zhang H, Yang C, Wei Z, Chen W, Du X, Liu J. Modulating Neuro-Immune-Induced Macrophage Polarization With Topiramate Attenuates Experimental Abdominal Aortic Aneurysm. Front Pharmacol 2020; 11:565461. [PMID: 32982758 PMCID: PMC7485436 DOI: 10.3389/fphar.2020.565461] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/12/2020] [Indexed: 12/14/2022] Open
Abstract
The development of abdominal aortic aneurysm (AAA) is attributed to psychological and physical factors. Topiramate, which is an agonist of the GABAA receptor, makes contributions to neuronal disease and is partially involved in immune regulation, may be effective upon abdominal aortic aneurysm progression. We used experimental abdominal aortic aneurysm models: Angiotensin II (Ang II)–induced ApoE−/− male mice (Ang II/APOE model) in our study. In the Ang II/APOE model, all mice (n=64) were divided into four groups: sham group (PBS treatment), control group (Ang II treatment), low-dose group (Ang II + low-dose topiramate, 3 mg/day per mouse), and high-dose group (Ang II + high-dose topiramate, 6 mg/day per mouse). All treatments began on the day after surgery. Moreover, collected tissues and cultured cell were used for histology and biochemical examination. In vitro, the effects of topiramate on bone marrow-derived macrophage stimulated by LPS were investigated. Our data implied that topiramate treatment significantly promoted macrophages preservation and conversion of M1 to M2 macrophage phenotypes in vivo and in vitro. Accordingly, proinflammatory activities mediated by the M1 macrophages were decreased and the repair process mediated by M2 macrophages was enhanced. The low-dose and high-dose groups had abdominal aortic aneurysm incidences of 50% and 37.5%, respectively, compared with 75% in the control group. Topiramate, a promising drug for the psychological disease, that target neuro-immune-induced macrophage polarization may attenuate experimental abdominal aortic aneurysm progression.
Collapse
Affiliation(s)
- Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Xiao
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanlei Yang
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanjie Wei
- Department of Thyroid and Breast Surgery, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Weiqiang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinling Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
49
|
Ma D, Zheng B, Liu HL, Zhao YB, Liu X, Zhang XH, Li Q, Shi WB, Suzuki T, Wen JK. Klf5 down-regulation induces vascular senescence through eIF5a depletion and mitochondrial fission. PLoS Biol 2020; 18:e3000808. [PMID: 32817651 PMCID: PMC7462304 DOI: 10.1371/journal.pbio.3000808] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 09/01/2020] [Accepted: 07/31/2020] [Indexed: 01/10/2023] Open
Abstract
Although dysregulation of mitochondrial dynamics has been linked to cellular senescence, which contributes to advanced age-related disorders, it is unclear how Krüppel-like factor 5 (Klf5), an essential transcriptional factor of cardiovascular remodeling, mediates the link between mitochondrial dynamics and vascular smooth muscle cell (VSMC) senescence. Here, we show that Klf5 down-regulation in VSMCs is correlated with rupture of abdominal aortic aneurysm (AAA), an age-related vascular disease. Mice lacking Klf5 in VSMCs exacerbate vascular senescence and progression of angiotensin II (Ang II)-induced AAA by facilitating reactive oxygen species (ROS) formation. Klf5 knockdown enhances, while Klf5 overexpression suppresses mitochondrial fission. Mechanistically, Klf5 activates eukaryotic translation initiation factor 5a (eIF5a) transcription through binding to the promoter of eIF5a, which in turn preserves mitochondrial integrity by interacting with mitofusin 1 (Mfn1). Accordingly, decreased expression of eIF5a elicited by Klf5 down-regulation leads to mitochondrial fission and excessive ROS production. Inhibition of mitochondrial fission decreases ROS production and VSMC senescence. Our studies provide a potential therapeutic target for age-related vascular disorders.
Collapse
Affiliation(s)
- Dong Ma
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - He-liang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Yong-bo Zhao
- Department of Cardiac surgery, the Fourth Hospital of Hebei Medical University, Shi Jiazhuang, China
| | - Xiao Liu
- Department of Cardiac surgery, the Fourth Hospital of Hebei Medical University, Shi Jiazhuang, China
| | - Xin-hua Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Qiang Li
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Wei-bo Shi
- Department of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| | - Toru Suzuki
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Jin-kun Wen
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
50
|
Qin Y, Zheng B, Yang GS, Yang HJ, Zhou J, Yang Z, Zhang XH, Zhao HY, Shi JH, Wen JK. Salvia miltiorrhiza-Derived Sal-miR-58 Induces Autophagy and Attenuates Inflammation in Vascular Smooth Muscle Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:492-511. [PMID: 32679544 PMCID: PMC7360890 DOI: 10.1016/j.omtn.2020.06.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
Autophagy is associated with the cytoprotection of physiological processes against inflammation and oxidative stress. Salvia miltiorrhiza possesses cardiovascular protective actions and has powerful anti-oxidative and anti-inflammatory effects; however, whether and how Salvia miltiorrhiza-derived microRNAs (miRNAs) protect vascular smooth muscle cells (VSMCs) by inducing autophagy across species are unknown. We first screened and identified Sal-miR-58 from Salvia miltiorrhiza as a natural autophagy inducer. Synthetic Sal-miR-58 suppresses chronic angiotensin II (Ang II) infusion-induced abdominal aortic aneurysm (AAA) formation in mice, as well as induces autophagy in VSMCs and attenuates the inflammatory response elicited by Ang II in vivo and in vitro. Mechanistically, Sal-miR-58 downregulates Krüppel-like factor 3 (KLF3) expression through direct binding to the 3' UTR of KLF3, which in turn relieves KLF3 repression of E3 ubiquitin ligase neural precursor cell-expressed developmentally downregulated 4-like (NEDD4L) expression, whereas NEDD4L upregulation increases the ubiquitination and degradation of the platelet isoform of phosphofructokinase (PFKP), subsequently leading to a decrease in the activation of Akt/mammalian target of rapamycin (mTOR) signaling and facilitating VSMC autophagy induced by Sal-miR-58 in the context of chronic Ang II stimulation and aneurysm formation. Our results provide the first evidence that plant-derived Sal-miR-58 induces autophagy and attenuates inflammation in VSMCs through cross-species modulation of the KLF3/NEDD4L/PFKP regulatory pathway.
Collapse
Affiliation(s)
- Yan Qin
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China; Department of Central Laboratory, Affiliated Hospital of Hebei University, Baoding 071000, China; Department of Life Science and Green Development, Hebei University, Baoding 071000, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Gao-Shan Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China; Department of Biochemistry and Molecular Biology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Hao-Jie Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Jing Zhou
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China; Department of Endocrine, The Second Hospital of Hebei Medical University, Shijiazhuang 050005, China
| | - Zhan Yang
- Department of Science and Technology, The Second Hospital of Hebei Medical University, Shijiazhuang 050005, China
| | - Xin-Hua Zhang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Hong-Ye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Jian-Hong Shi
- Department of Central Laboratory, Affiliated Hospital of Hebei University, Baoding 071000, China; Department of Life Science and Green Development, Hebei University, Baoding 071000, China
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|